首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 281 毫秒
1.
The TAM receptors—Tyro3, Axl, and Mer—comprise a unique family of receptor tyrosine kinases, in that as a group they play no essential role in embryonic development. Instead, they function as homeostatic regulators in adult tissues and organ systems that are subject to continuous challenge and renewal throughout life. Their regulatory roles are prominent in the mature immune, reproductive, hematopoietic, vascular, and nervous systems. The TAMs and their ligands—Gas6 and Protein S—are essential for the efficient phagocytosis of apoptotic cells and membranes in these tissues; and in the immune system, they act as pleiotropic inhibitors of the innate inflammatory response to pathogens. Deficiencies in TAM signaling are thought to contribute to chronic inflammatory and autoimmune disease in humans, and aberrantly elevated TAM signaling is strongly associated with cancer progression, metastasis, and resistance to targeted therapies.The name of the TAM family is derived from the first letter of its three constituents—Tyro3, Axl, and Mer (Prasad et al. 2006). As detailed in Figure 1, members of this receptor tyrosine kinase (RTK) family were independently identified by several different groups and appear in the early literature under multiple alternative names. However, Tyro3, Axl, and Mer (officially c-Mer or MerTK for the protein, Mertk for the gene) have now been adopted as the NCBI designations. The TAMs were first grouped into a distinct RTK family (the Tyro3/7/12 cluster) in 1991, through PCR cloning of their kinase domains (Lai and Lemke 1991). The isolation of full-length cDNAs for Axl (O''Bryan et al. 1991), Mer (Graham et al. 1994), and Tyro3 (Lai et al. 1994) confirmed their segregation into a structurally distinctive family of orphan RTKs (Manning et al. 2002b). The two ligands that bind and activate the TAMs—Gas6 and Protein S (Pros1)—were identified shortly thereafter (Ohashi et al. 1995; Stitt et al. 1995; Mark et al. 1996; Nagata et al. 1996).Open in a separate windowFigure 1.TAM receptors and ligands. The TAM receptors (red) are Tyro3 (Lai and Lemke 1991; Lai et al. 1994)—also designated Brt (Fujimoto and Yamamoto 1994), Dtk (Crosier et al. 1994), Rse (Mark et al. 1994), Sky (Ohashi et al. 1994), and Tif (Dai et al. 1994); Axl (O''Bryan et al. 1991)—also designated Ark (Rescigno et al. 1991), Tyro7 (Lai and Lemke 1991), and Ufo (Janssen et al. 1991); and Mer (Graham et al. 1994)—also designated Eyk (Jia and Hanafusa 1994), Nyk (Ling and Kung 1995), and Tyro12 (Lai and Lemke 1991). The TAMs are widely expressed by cells of the mature immune, nervous, vascular, and reproductive systems. The TAM ligands (blue) are Gas6 and Protein S (Pros1). The carboxy-terminal SHBG domains of the ligands bind to the immunoglobulin (Ig) domains of the receptors, induce dimerization, and activate the TAM tyrosine kinases. When γ-carboxylated in a vitamin-K-dependent reaction, the amino-terminal Gla domains of the dimeric ligands bind to the phospholipid phosphatidylserine expressed on the surface on an apposed apoptotic cell or enveloped virus. See text for details. (From Lemke and Burstyn-Cohen 2010; adapted, with permission, from the authors.)Subsequent progress on elucidating the biological roles of the TAM receptors was considerably slower and ultimately required the derivation of mouse loss-of-function mutants (Camenisch et al. 1999; Lu et al. 1999). The fact that Tyro3−/−, Axl−/−, and Mer−/− mice are all viable and fertile permitted the generation of a complete TAM mutant series that included all possible double mutants and even triple mutants that lack all three receptors (Lu et al. 1999). Remarkably, these Tyro3−/−Axl−/−Mer−/− triple knockouts (TAM TKOs) are viable, and for the first 2–3 wk after birth, superficially indistinguishable from their wild-type counterparts (Lu et al. 1999). Because many RTKs play essential roles in embryonic development, even single loss-of-function mutations in RTK genes often result in an embryonic-lethal phenotype (Gassmann et al. 1995; Lee et al. 1995; Soriano 1997; Arman et al. 1998). The postnatal viability of mice in which an entire RTK family is ablated completely—the TAM TKOs can survive for more than a year (Lu et al. 1999)—is therefore highly unusual. Their viability notwithstanding, the TAM mutants go on to develop a plethora of phenotypes, some of them debilitating (Camenisch et al. 1999; Lu et al. 1999; Lu and Lemke 2001; Scott et al. 2001; Duncan et al. 2003; Prasad et al. 2006). Almost without exception, these phenotypes are degenerative in nature and reflect the loss of TAM signaling activities in adult tissues that are subject to regular challenge, renewal, and remodeling. These activities are the subject of this review.  相似文献   

2.
RET (rearranged during transfection) is a receptor tyrosine kinase involved in the development of neural crest derived cell lineages, kidney, and male germ cells. Different human cancers, including papillary and medullary thyroid carcinomas, lung adenocarcinomas, and myeloproliferative disorders display gain-of-function mutations in RET. Accordingly, RET protein has become a promising molecular target for cancer treatment.The human RET (rearranged during transfection) gene maps on 10q11.2 and is composed of 21 exons spanning a region of 55,000 bp. It encodes a single-pass trans-membrane protein, RET, that belongs to the receptor tyrosine kinase (RTK) family (Pasini et al. 1995). The RET extracellular segment contains four cadherin-like domains, followed by a domain containing cysteine residues involved in the formation of intramolecular disulfide bonds (Fig. 1A) (Anders et al. 2001; Airaksinen and Saarma 2002). RET protein is highly glycosylated and N-glycosylation is necessary for its transport to the cell surface. Only the fully mature glycosylated 170 kDa RET protein isoform is exposed to the extracellular compartment, whereas the mannose-rich 150 kDa isoform is confined to the Golgi (Takahashi et al. 1993; Carlomagno et al. 1996). The transmembrane segment is composed of 22 amino acids, among which S649 and S653 mediate self-association and dimerization of RET, possibly via formation of inter-molecular hydrogen bonding (Kjaer et al. 2006). The intracellular portion of RET contains the tyrosine kinase domain split into two subdomains by the insertion of 27 amino acids. The RET COOH-terminal tail varies in length as a result of alternative splicing of the 3′ end (carboxy terminal with respect to glycine 1063), generating three different isoforms that contain 9 (RET9), 43 (RET43), or 51 (RET51) amino acids (Myers et al. 1995). RET9 and RET51 are the most abundant isoforms, and they activate similar signaling pathways through interaction with diverse protein complexes, and may exert a differential role in development (Fig. 1A) (de Graaff et al. 2001).Open in a separate windowFigure 1.Illustration of the mechanisms of activation of wild-type (wt) RET and RET-derived oncoproteins. (A) Wild-type RET activation is mediated by ligand (GFL)-induced dimerization; ligand binding to RET is not direct and mediated by GFR-α coreceptors (not shown); major RET autophosphorylation sites and downstream signaling pathways are indicated. RET extracellular cadherin-like domains are represented in red. The split intracellular RET tyrosine kinase domain, as well as the three alternative carboxy-terminal RET tails, are also depicted. (B) RET/PTC activation is mediated by coiled-coil-induced dimerization (left); activation of RET cysteine mutants associated with MEN2A or FMTC is mediated by disulfide bonds-mediated dimerization (right).RET shows several autophosphorylation sites (Fig. 1A) (Liu et al. 1996; Kawamoto et al. 2004). RET tyrosine 1062 (Y1062) functions as a multidocking site for signaling molecules containing a phosphotyrosine-binding (PTB) domain (Asai et al. 1996). Phospho-Y1062 binding proteins include SHC, N-SHC (RAI), FRS2, IRS1/2, DOK1, and DOK4/5 that, in turn, contribute to the activation of RAS-MAPK (mitogen-activated protein kinases) and PI3K (phosphatidyl inositol 3 kinase)-AKT pathways. Y1096, specific to the RET51 splicing variant, couples to the PI3K-AKT and RAS-MAPK pathways, as well. These signaling cascades mediate RET-dependent cell survival, proliferation, and motility (Alberti et al. 1998; Murakami et al. 1999; Segouffin-Cariou and Billaud 2000; Melillo et al. 2001a,b; Schuetz et al. 2004). Y905 is located in the activation loop of the RET kinase and its phosphorylation is associated with RET kinase activation (Knowles et al. 2006). Finally, Y981 and Y1015 have been shown to be coupled to important signaling molecules such as SRC and PLC-γ, respectively (Borrello et al. 1996; Encinas et al. 2004).RET is the receptor for a group of neurotrophic growth factors that belong to the glial cell line-derived neurotrophic factor (GDNF) family (GFLs, GDNF family ligands), namely, GDNF, Neurturin (NRT), Artemin (ART), and Persephin (PSF) (Airaksinen and Saarma 2002). GFLs mediate RET protein dimerization and activation (Fig. 1A). GFLs are presented to RET by GPI (glycosylphosphatidylinositol)-anchored coreceptors, called GFR-α (GDNF family receptor α 1-4). Differential tissue expression dictates the specificity of action displayed by alternative GLF-GFR-α pairs during development and adult life (Baloh et al. 2000; Airaksinen and Saarma 2002).Together with other membrane (DCC and p75NTR) or nuclear (androgen receptor, AR) receptors, RET belongs to the family of so-called “dependence” receptors (Mehlen and Bredesen 2011). In the absence of ligand, RET exerts a proapoptotic activity, that is blocked on ligand stimulation (Bordeaux et al. 2000). Such pro-apoptotic activity is RET kinase-independent and mediated by cleavage of RET cytosolic portion by caspase-3, which, in turn, releases a carboxy-terminal RET peptide that is able to induce cell death (Bordeaux et al. 2000). It is feasible that such activity is important for RET developmental function, because it may control migration of RET-expressing cells by limiting survival of cells that move beyond ligand availability (Bordeaux et al. 2000; Cañibano et al. 2007). Whether modulation of this function is also important for RET-associated diseases is still unknown. However, it is interesting to note that a cancer-associated RET mutant (RET-C634R, see below) does not exert cleavage-dependent proapoptotic effects, whereas RET mutants associated with defective development (Hirschsprung disease, see below) exert strong proapoptotic activity that is refractory to modulation by ligand (Bordeaux et al. 2000).RET is expressed in enteric ganglia, adrenal medulla chromaffin cells, thyroid C cells, sensory and autonomic ganglia of the peripheral nervous system, a subset of central nervous system nuclei, developing kidney and testis germ cells (Manié et al. 2001; de Graaff et al. 2001). RET null mice display impaired development of superior cervical ganglia and enteric nervous system, kidney agenesia, reduction of thyroid C cells, and impaired spermatogenesis (Manié et al. 2001). Accordingly, individuals with germline loss-of-function mutations of RET are affected by intestinal aganglionosis causing congenital megacolon (Hirschsprung disease) (Brooks et al. 2005). RET loss-of-function mutations have also been identified in congenital anomalies of kidney and urinary tract (CAKUT), either isolated or in combination with Hirschsprung disease (Jain 2009).Several genetic alterations convert RET into a dominantly transforming oncogene. This review will describe RET-derived oncogenes that are associated with different types of human neoplasia (Fig. 1B).  相似文献   

3.
The eukaryotic cytoskeleton evolved from prokaryotic cytomotive filaments. Prokaryotic filament systems show bewildering structural and dynamic complexity and, in many aspects, prefigure the self-organizing properties of the eukaryotic cytoskeleton. Here, the dynamic properties of the prokaryotic and eukaryotic cytoskeleton are compared, and how these relate to function and evolution of organellar networks is discussed. The evolution of new aspects of filament dynamics in eukaryotes, including severing and branching, and the advent of molecular motors converted the eukaryotic cytoskeleton into a self-organizing “active gel,” the dynamics of which can only be described with computational models. Advances in modeling and comparative genomics hold promise of a better understanding of the evolution of the self-organizing cytoskeleton in early eukaryotes, and its role in the evolution of novel eukaryotic functions, such as amoeboid motility, mitosis, and ciliary swimming.The eukaryotic cytoskeleton organizes space on the cellular scale and this organization influences almost every process in the cell. Organization depends on the mechanochemical properties of the cytoskeleton that dynamically maintain cell shape, position organelles, and macromolecules by trafficking, and drive locomotion via actin-rich cellular protrusions, ciliary beating, or ciliary gliding. The eukaryotic cytoskeleton is best described as an “active gel,” a cross-linked network of polymers (gel) in which many of the links are active motors that can move the polymers relative to each other (Karsenti et al. 2006). Because prokaryotes have only cytoskeletal polymers but lack motor proteins, this “active gel” property clearly sets the eukaryotic cytoskeleton apart from prokaryotic filament systems.Prokaryotes contain elaborate systems of several cytomotive filaments (Löwe and Amos 2009) that share many structural and dynamic features with eukaryotic actin filaments and microtubules (Löwe and Amos 1998; van den Ent et al. 2001). Prokaryotic cytoskeletal filaments may trace back to the first cells and may have originated as higher-order assemblies of enzymes (Noree et al. 2010; Barry and Gitai 2011). These cytomotive filaments are required for the segregation of low copy number plasmids, cell rigidity and cell-wall synthesis, cell division, and occasionally the organization of membranous organelles (Komeili et al. 2006; Thanbichler and Shapiro 2008; Löwe and Amos 2009). These functions are performed by dynamic filament-forming systems that harness the energy from nucleotide hydrolysis to generate forces either via bending or polymerization (Löwe and Amos 2009; Pilhofer and Jensen 2013). Although the identification of actin and tubulin homologs in prokaryotes is a major breakthrough, we are far from understanding the origin of the structural and dynamic complexity of the eukaryotic cytoskeleton.Advances in genome sequencing and comparative genomics now allow a detailed reconstruction of the cytoskeletal components present in the last common ancestor of eukaryotes. These studies all point to an ancestrally complex cytoskeleton, with several families of motors (Wickstead and Gull 2007; Wickstead et al. 2010) and filament-associated proteins and other regulators in place (Jékely 2003; Richards and Cavalier-Smith 2005; Rivero and Cvrcková 2007; Chalkia et al. 2008; Eme et al. 2009; Fritz-Laylin et al. 2010; Eckert et al. 2011; Hammesfahr and Kollmar 2012). Genomic reconstructions and comparative cell biology of single-celled eukaryotes (Raikov 1994; Cavalier-Smith 2013) allow us to infer the cellular features of the ancestral eukaryote. These analyses indicate that amoeboid motility (Fritz-Laylin et al. 2010; although, see Cavalier-Smith 2013), cilia (Cavalier-Smith 2002; Mitchell 2004; Jékely and Arendt 2006; Satir et al. 2008), centrioles (Carvalho-Santos et al. 2010), phagocytosis (Cavalier-Smith 2002; Jékely 2007; Yutin et al. 2009), a midbody during cell division (Eme et al. 2009), mitosis (Raikov 1994), and meiosis (Ramesh et al. 2005) were all ancestral eukaryotic cellular features. The availability of functional information from organisms other than animals and yeasts (e.g., Chlamydomonas, Tetrahymena, Trypanosoma) also allow more reliable inferences about the ancestral functions of cytoskeletal components (i.e., not only their ancestral presence or absence) and their regulation (Demonchy et al. 2009; Lechtreck et al. 2009; Suryavanshi et al. 2010).The ancestral complexity of the cytoskeleton in eukaryotes leaves a huge gap between prokaryotes and the earliest eukaryote we can reconstruct (provided that our rooting of the tree is correct) (Cavalier-Smith 2013). Nevertheless, we can attempt to infer the series of events that happened along the stem lineage, leading to the last common ancestor of eukaryotes. Meaningful answers will require the use of a combination of gene family history reconstructions (Wickstead and Gull 2007; Wickstead et al. 2010), transition analyses (Cavalier-Smith 2002), and computer simulations relevant to cell evolution (Jékely 2008).  相似文献   

4.
Fibronectin (FN) is a multidomain protein with the ability to bind simultaneously to cell surface receptors, collagen, proteoglycans, and other FN molecules. Many of these domains and interactions are also involved in the assembly of FN dimers into a multimeric fibrillar matrix. When, where, and how FN binds to its various partners must be controlled and coordinated during fibrillogenesis. Steps in the process of FN fibrillogenesis including FN self-association, receptor activities, and intracellular pathways have been under intense investigation for years. In this review, the domain organization of FN including the extra domains and variable region that are controlled by alternative splicing are described. We discuss how FN–FN and cell–FN interactions play essential roles in the initiation and progression of matrix assembly using complementary results from cell culture and embryonic model systems that have enhanced our understanding of this process.As a ubiquitous component of the extracellular matrix (ECM), fibronectin (FN) provides essential connections to cells through integrins and other receptors and regulates cell adhesion, migration, and differentiation. FN is secreted as a large dimeric glycoprotein with subunits that range in size from 230 kDa to 270 kDa (Mosher 1989; Hynes 1990). Variation in subunit size depends primarily on alternative splicing. FN was first isolated from blood more than 60 years ago (Edsall 1978), and this form is called plasma FN. The other major form, called cellular FN, is abundant in the fibrillar matrices of most tissues. Although FN is probably best known for promoting attachment of cells to surfaces, this multidomain protein has many interesting structural features and functional roles beyond cell adhesion.FN is composed of three different types of modules termed type I, II, and III repeats (Fig. 1) (Petersen et al. 1983; Hynes 1990). These repeats have distinct structures. Although the conformations of type I and type II repeats are maintained by pairs of intramodule disulfide bonds, the type III repeat is a 7-stranded β-barrel structure that lacks disulfide bonds (Main et al. 1992; Leahy et al. 1996, 1992) and, therefore, can undergo conformational changes. FN type III repeats are widely distributed among animal, bacterial, and plant proteins and are found in both extracellular and intracellular proteins (Bork and Doolittle 1992; Tsyguelnaia and Doolittle 1998).Open in a separate windowFigure 1.FN domain organization and isoforms. Each FN monomer has a modular structure consisting of 12 type I repeats (cylinders), 2 type II repeats (diamonds), and 15 constitutive type III repeats (hexagons). Two additional type III repeats (EIIIA and EIIIB, green) are included or omitted by alternative splicing. The third region of alternative splicing, the V region (green box), is included (V120), excluded (V0), or partially included (V95, V64, V89). Sets of modules comprise domains for binding to other extracellular molecules as indicated. Domains required for fibrillogenesis are in red: the assembly domain (repeats I1-5) binds FN, III9-10 contains the RGD and synergy sequences for integrin binding, and the carboxy-terminal cysteines form the disulfide-bonded FN dimer (‖). The III1-2 domain (light red) has two FN binding sites that are important for fibrillogenesis. The amino-terminal 70-kDa fragment contains assembly and gelatin-binding domains and is routinely used in FN binding and matrix assembly studies.Sets of adjacent modules form binding domains for a variety of proteins and carbohydrates (Fig. 1). ECM proteins, including FN, bind to cells via integrin receptors, αβ heterodimers with two transmembrane subunits (Hynes 2002). FN-binding integrins have specificity for one of the two cell-binding sites within FN, either the RGD-dependent cell-binding domain in III10 (Pierschbacher and Ruoslahti 1984) or the CS1 segment of the alternatively spliced V region (IIICS) (Wayner et al. 1989; Guan and Hynes 1990). Some integrins require a synergy sequence in repeat III9 for maximal interactions with FN (Aota et al. 1994; Bowditch et al. 1994). Another family of cell surface receptors is the syndecans, single-chain transmembrane proteoglycans (Couchman 2010). Syndecans use their glycosaminoglycan (GAG) chains to interact with FN at its carboxy-terminal heparin-binding (HepII) domain (Fig. 1) (Saunders and Bernfield 1988; Woods et al. 2000), which binds to heparin, heparan sulfate, and chondroitin sulfate GAGs (Hynes 1990; Barkalow and Schwarzbauer 1994). Syndecan binding to the HepII domain enhances integrin-mediated cell spreading and intracellular signaling, suggesting that syndecans act as coreceptors with integrins in cell–FN binding (Woods and Couchman 1998; Morgan et al. 2007).A major site for FN self-association is within the amino-terminal assembly domain spanning the first five type I repeats (I1-5) (Fig. 1) (McKeown-Longo and Mosher 1985; McDonald et al. 1987; Schwarzbauer 1991b; Sottile et al. 1991). This domain plays an essential role in FN fibrillogenesis. As a major blood protein, FN interacts with fibrin during blood coagulation, also using the I1-5 domain (Mosher 1989; Hynes 1990). As fibrin polymerizes, factor XIII transglutaminase covalently cross-links glutamine residues near the amino terminus of FN to fibrin α chains (Mosher 1975; Corbett et al. 1997). The amino-terminal domain has multiple binding partners in addition to FN and fibrin; these include heparin, S. aureus, and other bacteria, thrombospondin-1, and tenascin-C (Hynes 1990; Ingham et al. 2004; Schwarz-Linek et al. 2006). Adjacent to this domain is the gelatin/collagen-binding domain composed of type I and type II modules (Ingham et al. 1988). This domain also binds to tissue transglutaminase (Radek et al. 1993) and fibrillin-1 (Sabatier et al. 2009). Within the 15 type III repeats reside several FN binding sites that interact with the amino-terminal assembly domain as well as three sites of alternative splicing that generate multiple isoforms. At the carboxyl terminus is a pair of cysteine residues that form the FN dimer through antiparallel disulfide bonds (Hynes 1990). This dimerization may be facilitated by disulfide isomerase activity located in the last set of type I repeats (Langenbach and Sottile 1999).The diverse set of binding domains provides FN with the ability to interact simultaneously with other FN molecules, other ECM components (e.g., collagens and proteoglycans), cell surface receptors, and extracellular enzymes (Pankov and Yamada 2002; Fogelgren et al. 2005; Hynes 2009; Singh et al. 2010). Multitasking by FN probably underlies its essential role during embryogenesis (George et al. 1993). Furthermore, FN''s interactions can be modulated by exposure or sequestration of its binding sites within matrix fibrils, through the presence of ECM proteins that bind to FN, or through variation in structure by alternative splicing.  相似文献   

5.
Epithelial cell–cell junctions are formed by apical adherens junctions (AJs), which are composed of cadherin adhesion molecules interacting in a dynamic way with the cortical actin cytoskeleton. Regulation of cell–cell junction stability and dynamics is crucial to maintain tissue integrity and allow tissue remodeling throughout development. Actin filament turnover and organization are tightly controlled together with myosin-II activity to produce mechanical forces that drive the assembly, maintenance, and remodeling of AJs. In this review, we will discuss these three distinct stages in the lifespan of cell–cell junctions, using several developmental contexts, which illustrate how mechanical forces are generated and transmitted at junctions, and how they impact on the integrity and the remodeling of cell–cell junctions.Cell–cell junction formation and remodeling occur repeatedly throughout development. Epithelial cells are linked by apical adherens junctions (AJs) that rely on the cadherin-catenin-actin module. Cadherins, of which epithelial E-cadherin (E-cad) is the most studied, are Ca2+-dependent transmembrane adhesion proteins forming homophilic and heterophilic bonds in trans between adjacent cells. Cadherins and the actin cytoskeleton are mutually interdependent (Jaffe et al. 1990; Matsuzaki et al. 1990; Hirano et al. 1992; Oyama et al. 1994; Angres et al. 1996; Orsulic and Peifer 1996; Adams et al. 1998; Zhang et al. 2005; Pilot et al. 2006). This has long been attributed to direct physical interaction of E-cad with β-catenin (β-cat) and of α-catenin (α-cat) with actin filaments (for reviews, see Gumbiner 2005; Leckband and Prakasam 2006; Pokutta and Weis 2007). Recently, biochemical and protein dynamics analyses have shown that such a link may not exist and that instead, a constant shuttling of α-cat between cadherin/β-cat complexes and actin may be key to explain the dynamic aspect of cell–cell adhesion (Drees et al. 2005; Yamada et al. 2005). Regardless of the exact nature of this link, several studies show that AJs are indeed physically attached to actin and that cadherins transmit cortical forces exerted by junctional acto-myosin networks (Costa et al. 1998; Sako et al. 1998; Pettitt et al. 2003; Dawes-Hoang et al. 2005; Cavey et al. 2008; Martin et al. 2008; Rauzi et al. 2008). In addition, physical association depends in part on α-cat (Cavey et al. 2008) and additional intermediates have been proposed to represent alternative missing links (Abe and Takeichi 2008) (reviewed in Gates and Peifer 2005; Weis and Nelson 2006). Although further work is needed to address the molecular nature of cadherin/actin dynamic interactions, association with actin is crucial all throughout the lifespan of AJs. In this article, we will review our current understanding of the molecular mechanisms at work during three different developmental stages of AJs biology: assembly, stabilization, and remodeling, with special emphasis on the mechanical forces controlling AJs integrity and development.  相似文献   

6.
7.
Proteins to be secreted are transported from the endoplasmic reticulum (ER) to the Golgi apparatus. The transport of these proteins requires the localization and activity of proteins that create ER exit sites, coat proteins to collect cargo and to reshape the membrane into a transport container, and address labels—SNARE proteins—to target the vesicles specifically to the Golgi apparatus. In addition some proteins may need export chaperones or export receptors to enable their exit into transport vesicles. ER export factors, SNAREs, and misfolded Golgi-resident proteins must all be retrieved from the Golgi to the ER again. This retrieval is also part of the organellar homeostasis pathway essential to maintaining the identity of the ER and of the Golgi apparatus. In this review, I will discuss the different processes in retrograde transport from the Golgi to the ER and highlight the mechanistic insights we have obtained in the last couple of years.Proteins that are exposed at the plasma membrane or populate a membrane-bounded organelle are synthesized into the endoplasmic reticulum (ER). In the ER, the folding of these proteins takes place and posttranslational modifications such as N-glycosylation and disulfide bridge formation occur. Upon adopting a suitable, often correct, conformation, proteins destined to locations beyond the ER are concentrated at so-called ER exit sites (ERES) and incorporated into nascent COPII-coated vesicles. These COPII vesicles eventually bud off the ER membrane and are transported to the Golgi (in yeast, Drosophila, and C. elegans) or the ER-Golgi intermediate compartment (in mammalian cells) (Schweizer et al. 1990; Kondylis and Rabouille 2003; Spang 2009; Witte et al. 2011).It is assumed that the vesicle coat is at least partially destabilized through the hydrolysis of GTP by the small GTPase Sar1 (Oka and Nakano 1994; Springer et al. 1999). However, some of the destabilized coat components have to stay on the vesicle until it has reached the Golgi apparatus because coat components participate in the recognition and the tethering process (Barlowe 1997; Cai et al. 2007; Lord et al. 2011; Zong et al. 2012). Subsequently, SNARE proteins on the vesicles (v-SNAREs) zipper up with cognate SNAREs on the Golgi (target SNAREs, t-SNAREs) to drive membrane fusion (Hay et al. 1998; Cao and Barlowe 2000; Parlati et al. 2002). The content of the ER-derived COPII vesicles is thereby released into the lumen of the cis-cisterna of the Golgi apparatus. Most proteins will continue their journey through the Golgi apparatus and encounter further modifications such as extension of the glycosylation tree or lipidation. However, some proteins, especially those involved in the fusion process, i.e., the v-SNAREs or proteins that act as export factors of the ER, such as Vma21, which is essential for export of the correctly folded and assembled V0 sector of the V-ATPase, need to be recycled back to the ER for another round of transport (Ballensiefen et al. 1998; Malkus et al. 2004). Moreover, cis-Golgi proteins are returned to the ER for quality/functional control (Todorow et al. 2000; Sato et al. 2004; Valkova et al. 2011). Finally, some ER-resident proteins, such as the ER Hsp70 chaperone BiP/Kar2, can escape the ER, but are captured at the cis-Golgi by the H/KDEL receptor Erd2 and returned to the ER (Lewis et al. 1990; Semenza et al. 1990; Aoe et al. 1997).Unfortunately, the retrograde transport route is also hijacked by toxins. For example, endocytosed cholera toxin subunit A contains a KDEL sequence and can thereby exploit the system to access the ER (Majoul et al. 1996, 1998). From there, it is retro-translocated into the cytoplasm where it can exert its detrimental function.  相似文献   

8.
9.
Nodes of Ranvier and axon initial segments of myelinated nerves, sites of cell–cell contact in early embryos and epithelial cells, and neuromuscular junctions of skeletal muscle all perform physiological functions that depend on clustering of functionally related but structurally diverse ion transporters and cell adhesion molecules within microdomains of the plasma membrane. These specialized cell surface domains appeared at different times in metazoan evolution, involve a variety of cell types, and are populated by distinct membrane-spanning proteins. Nevertheless, recent work has shown that these domains all share on their cytoplasmic surfaces a membrane skeleton comprised of members of the ankyrin and spectrin families. This review will summarize basic features of ankyrins and spectrins, and will discuss emerging evidence that these proteins are key players in a conserved mechanism responsible for assembly and maintenance of physiologically important domains on the surfaces of diverse cells.Spectrins are flexible rods 0.2 microns in length with actin-binding sites at each end (Shotton et al. 1979; Bennett et al. 1982) (Fig. 1A). Spectrins are assembled from α and β subunits, each comprised primarily of multiple copies of a 106-amino acid repeat (Speicher and Marchesi 1984). In addition to the canonical 106-residue repeat, β spectrins also have a carboxy-terminal pleckstrin homology domain (Zhang et al. 1995; Macias et al. 1994) and tandem amino-terminal calponin homology domains (Bañuelos et al. 1998), whereas α spectrins contain an Src homology domain 3 (SH3) site (Musacchio et al. 1992), a calmodulin-binding site (Simonovic et al. 2006), and EF hands (Travé et al. 1995) (Fig. 1A). Spectrin α and β subunits are assembled antiparallel and side-to-side into heterodimers, which in turn are associated head-to-head to form tetramers (Clarke 1971; Shotton et al. 1979; Davis and Bennett 1983) (Fig. 1A). In human erythrocytes, in which spectrin was first characterized (Marchesi and Steers 1968; Clarke 1971), actin oligomers containing 10–14 monomers are each linked to five to six spectrin tetramers by accessory proteins to form a geodesic domelike structure that has been resolved by electron microscopy (Byers and Branton 1985). The principal proteins at the spectrin–actin junction are protein 4.1, adducin, tropomyosin, tropomodulin, and dematin (Bennett and Baines 2001) (Open in a separate windowFigure 1.Domain structure and variants of spectrin and ankyrin proteins. (A) Molecular domains of spectrins: Two α spectrins and five β spectrins are shown. Spectrins are comprised of modular units called spectrin repeats (yellow). Other domains such as the ankyrin binding domain (purple), Src-homology domain 3 (SH3, blue), EF-hand domain (red), and calmodulin-binding domain (green) promote interactions with binding targets important for spectrin function. The pleckstrin homology domain (black) promotes association with the plasma membrane and the actin binding domain (grey) tethers the spectrin-based membrane skeleton to the underlying actin cytoskeleton. (B) The spectrin tetramer, the fundamental unit of the spectrin-based membrane skeleton. The spectrin repeat domains of α and β spectrin associate end-to-end to form heterodimers. Heterodimers associate laterally in an antiparallel fashion to form tetramers. The tetramers can then associate end-to-end to form extended macromolecules that link into a geodesic dome shape directly underneath the plasma membrane. (C) Molecular domains present in canonical ankyrins. The membrane binding domain of ankyrin isoforms (orange) is comprised of 24 ANK repeats. The spectrin binding domain (green-blue) allows ankyrins to coordinate integral membrane proteins to the membrane skeleton. The death domain (pink) is the most highly conserved domain. The regulatory domain (brown) is the most variable region of ankyrins. The regulatory domain interacts intramolecularly with the membrane binding domain to modulate ankyrin’s affinity for other binding partners. All ankyrins and spectrins are subject to alternative splicing, which further increases their functional diversity.

Table 1.

Binding partners of spectrin and ankyrins
Spectrin Binding Partners
AlphaBeta
Transporters/ion channels
EnNaC (sodium)
NHE2 (ammonium)
Membrane anchors
PI lipids
Band 4.1
Ankyrin
EAAT4 (glutamate)
Membrane receptors
NMDA receptor
Signaling
RACK-1
Signaling
HsSH3pb1
Calmodulin
Cytoskeleton/cellular transport
F-actin
Adducin
Dynactin
Ankyrin Binding Partners
Membrane BDSpectrin BDDDREG D
Ion channels:
Anion exchanger
Na+/K+ATPase
Voltage-gated
Na+ channels
Na+/Ca2+ Exchanger
KCNG2/3
Rh antigen
IP3 receptor
Ryanodine receptor
Cell adhesion molecules:
L1-CAMs
CD44
E-cadherin
Dystroglycan
Cellular transport:
Tubulin
Clathrin
SpectrinFasLHsp40
Obscurin
PP2A
Open in a separate windowSpectrin is coupled to the inner surface of the erythrocyte membrane primarily through association with ankyrin, which is in turn linked to the cytoplasmic domains of the anion exchanger (Bennett 1978; Bennett and Stenbuck 1979a,b) and Rh/RhAG ammonium transporter (Nicolas et al. 2003). The spectrin-based membrane skeleton and its connections through ankyrin to membrane-spanning proteins are essential for survival of erythrocytes in the circulation, and mutations in these proteins result in hereditary hemolytic anemia (Bennett and Healy 2008). The ankyrin-binding sites of β spectrins 1–4 are located in the 15th spectrin repeat, which is folded identically to other repeats but has distinct surface-exposed residues (Davis et al. 2008; Ipsaro et al. 2009; Stabach et al. 2009) (Figs. 1A, A,2A).2A). Mammalian β-5 spectrin and its ortholog β-H spectrin in Drosophila and Caenorhabditis elegans are the only β spectrins lacking ankyrin-binding activity (Dubreuil et al. 1990; Thomas et al. 1998; McKeown et al. 1998; Stabach and Morrow 2000).Open in a separate windowFigure 2.Ankyrins and spectrins organize macromolecular complexes in diverse types of specialized membranes. (A) Ankyrin-G forms a complex with β-IV spectrin, neurofascin (a cell adhesion protein), and ion channels (KCNQ2/3 and voltage-gated sodium channel) at axon initial segments in Purkinje neurons. (B) In force buffering costameres of skeletal muscle, ankyrins -B and -G cooperate to target and stabilize key components of the dystroglycoprotein complex. At the membrane, ankyrin-G binds to dystrophin and β-dystroglycan. (C) In cardiomyocyte transverse tubules, ankyrins -B and -G coordinate separate microdomains. Ankyrin-B binds Na+/K+ ATPase, Na+/Ca2+ exchanger (NCX-1), and the inositol triphosphate receptor (IP3R). Ankyrin-G forms a complex with Nav1.5 and spectrin. (D) Ankyrin-G in epithelial lateral membrane assembly. Ankyrin-G binds to E-cadherin, β-2 spectrin, and the Na+/K+ ATPase. Spectrins are connected via F-actin bridges bound to α/γ adducin and tropomodulin.Ankyrin interacts with β spectrins through a ZU5 domain (Mohler et al. 2004a; Kizhatil et al. 2007a; Ipsaro et al. 2009) (Fig. 1B), and with most of its membrane partners through ANK repeats (Bennett and Baines 2001) (Fig. 2C,D). In addition, ankyrins have a highly conserve “death domain” and a carboxy-terminal regulatory domain (see the following discussion). The 24 ANK repeats are stacked in a superhelical array to form a solenoid (Michaely et al. 2002). Interestingly, the ANK repeat stack behaves like a reversible spring when stretched by atomic force microscopy, and may function in mechano-coupling in tissues such as the heart (Lee et al. 2006). ANK repeats are components of many proteins and participate in highly diverse protein interactions (Mosavi et al. 2004) (Fig. 2C). This versatile motif currently is being exploited using designed ANK repeat proteins (DARPins) engineered to interact with specific ligands that can function as substitutes for antibodies (Stumpp and Amstutz 2007; Steiner et al. 2008).Spectrin and ankyrin family members are expressed in most, if not all, animal (metazoan) cells, but are not present in bacteria, plants, or fungi. Spectrins are believed to have evolved from an ancestral α-actinin containing calponin homology domains and two spectrin repeats but not other domains (Thomas et al. 1997; Pascual et al. 1997). Ankyrin repeats are expressed in all phyla, presumably because of a combination of evolutionary relationships and in cases of bacteria and viruses by horizontal gene transfer. However, the spectrin-binding domain of ankyrin is present only in metazoans (Fig 1B). It is possible that evolution of ankyrins and spectrins could have been one of the adaptations required for organization of cells into tissues in multicellular animals.The human spectrin family includes two α subunits and five β subunits, whereas Drosophila and C. elegans have a single α subunit and two β subunits (Bennett and Baines 2001). Vertebrate ankyrins are encoded by three genes: ankyrin-R (ANK1) (the isoform first characterized in erythrocytes and also present in a restricted distribution in brain and muscle), ankyrin-B (ANK2), and ankyrin-G (ANK3). Vertebrate ankyrins evolved from a single gene in early chordates (Cai and Zhang 2006). C. elegans ankyrin is encoded by a single gene termed unc-44 (Otsuka et al. 1995), whereas the Drosophila genome contains two ankyrin genes: ankyrin (Dubreuil and Yu 1994) and ankyrin2 (Bouley et al. 2000).Mammalian ankyrins -B and -G are co-expressed in most cells, although they have distinct functions (Mohler et al. 2002; Abdi et al. 2006). Ankyrins -B and -G are closely related in their ANK repeats, and spectrin-binding domains, but diverge in their carboxy-terminal regulatory domains. Regulatory domains are natively unstructured and extended (Abdi et al. 2006). These flexible domains engage in intramolecular interactions with the membrane-binding and spectrin-binding domains (Hall and Bennett 1987; Davis et al. 1992; Abdi et al. 2006) that modulate protein associations and provide functional diversity between otherwise conserved ankyrins.In addition to the standard versions of ankyrins and spectrin subunits depicted in Figure 1, many variants of these proteins are expressed with the addition and/or deletion of functional domains because of alternative splicing of pre-mRNAs. For example, β spectrins can lack PH domains (Hayes et al. 2000), and giant ankyrins have insertions of up to 2000 residues (Kordeli et al. 1995; Chan et al. 1993; Pielage et al. 2008; Koch et al. 2008), whereas other ankyrins lack either the entire membrane-binding domain (Hoock et al. 1997), or both membrane- and spectrin-binding domains (Zhou et al. 1997). The insertions in 440 kDa ankyrin-B and 480 kDa ankyrin-G (Fig. 1B) have an extended conformation that potentially could have specialized roles in connections between the plasma membrane and cytoskeleton of axons where these giant ankyrins reside (Chan et al. 1993; Kordeli et al. 1995) (Fig. 1B). Interestingly, the inserted sequences in Drosophila giant ankyrins interact with microtubules at the presynaptic neuromuscular junction (Pielage et al. 2008) (see the following section).  相似文献   

10.
11.
Since its first visualization in 1898, the Golgi has been a topic of intense morphological research. A typical mammalian Golgi consists of a pile of stapled cisternae, the Golgi stack, which is a key station for modification of newly synthesized proteins and lipids. Distinct stacks are interconnected by tubules to form the Golgi ribbon. At the entrance site of the Golgi, the cis-Golgi, vesicular tubular clusters (VTCs) form the intermediate between the endoplasmic reticulum and the Golgi stack. At the exit site of the Golgi, the trans-Golgi, the trans-Golgi network (TGN) is the major site of sorting proteins to distinct cellular locations. Golgi functioning can only be understood in light of its complex architecture, as was revealed by a range of distinct electron microscopy (EM) approaches. In this article, a general concept of mammalian Golgi architecture, including VTCs and the TGN, is described.In 1898 Camillo Golgi was the first to visualize, describe, and ultimately name the Golgi complex. Using a histochemical impregnation method causing the reduction and deposition of silver, he defined the Golgi in neuronal cells as a reticular apparatus stained by the “black reaction” (Golgi 1898). In the 1950s, the first ultrastructural images of the Golgi were revealed using the then newly developed electron microscope (EM) (Dalton 1954; Farquhar and Rinehart 1954; Sjostrand and Hanzon 1954; Dalton and Felix 1956), reviewed by Farquhar and Palade (1981). In 1961, the thiamine pyrophosphatase reaction developed by Novikoff and Goldfischer allowed cytochemical labeling of Golgi membranes, which revealed the ubiquitous cellular distribution of this organelle (Novikoff and Goldfischer 1961). In the many years of ultrastructural research that have followed, the visualization of the Golgi has gone hand-in-hand with the developing EM techniques.The intriguing structural complexity of the Golgi has made it one of the most photographed organelles in the cell. However, a full understanding of Golgi architecture is hard to deduce from the ultrathin (70–100 nm) sections used in standard transmission EM preparations. Rambourg and Clermont (1974) were the first to investigate the Golgi in three dimensions (3D), using stereoscopy (Rambourg 1974). In this approach a “thick” (150–200 nm), EM section is photographed at two distinct angles, after which the pairs of photographs are viewed with a stereoscope. Over the years, stereoscopy was applied to a variety of cells and has greatly contributed to our current understanding of Golgi architecture (Lindsey and Ellisman 1985; Rambourg and Clermont 1990; Clermont et al. 1994; Clermont et al. 1995). An alternative approach to study 3D structure is serial sectioning, by which a series of adjacent (serial) thin sections are collected. The Golgi can be followed throughout these sections and be constructed into a 3D model (Beams and Kessel 1968; Dylewski et al. 1984; Rambourg and Clermont 1990). In the nineties, 3D-EM was boosted by the introduction of high-voltage, dual axis 3D electron tomography (Ladinsky et al. 1999; Koster and Klumperman 2003; Marsh 2005; Marsh 2007; Noske et al. 2008), which allows the analysis of sections of up to 3–4 µm with a 4–6 nm resolution in the z-axis. The sections are photographed in a tilt series of different angles, which are reconstructed into a 3D tomogram that allows one to “look beyond” a given structure and reveals how it relates to other cellular compartments.Membranes with a similar appearance can differ in protein content and function. These differences are revealed by protein localization techniques. Therefore, in addition to the “classical” EM techniques providing ultrastructural details, EM methods that determine protein localization within the context of the cellular morphology have been crucial to further our understanding on the functional organization of the Golgi. For example, by enzyme-activity-based cytochemical staining the cis-to-trans-polarity in the distribution of Golgi glycosylation enzymes was discovered, reviewed by Farquhar and Palade (1981), which was key to understanding the functional organization of the Golgi stack in protein and lipid glycosylation. With the development of immunoEM methods, using antibodies, the need for enzyme activity for protein localization was overcome. This paved the way for the localization of a wide variety of proteins, such as the cytoplasmic coat complexes associated with the Golgi (Rabouille and Klumperman 2005).A logical next step in EM-based imaging of the Golgi would be to combine protein localization with 3D imaging, but this is technically challenging. A number of protocols enabling protein localization in 3D have recently been described (Trucco et al. 2004; Grabenbauer et al. 2005; Gaietta et al. 2006; Zeuschner et al. 2006; Meiblitzer-Ruppitsch et al. 2008), but these have only been applied in a limited manner to Golgi studies. Another approach that holds great potential for Golgi research is correlative microscopy (CLEM). Live cell imaging of fluorescent proteins has revolutionized cell biology by the real time visualization of dynamic events. However, live cell imaging does not reveal membrane complexity. By CLEM, live cells are first viewed by light microscopy and then prepared for EM (Mironov et al. 2008; van Rijnsoever et al. 2008). When coupled with the recent introduction of super resolution light microscopy techniques for real time imaging, the combination with EM for direct correlation with ultrastructural resolution has great potential (Hell 2009; Lippincott-Schwartz and Manley 2009).The 100th anniversary of the discovery of the Golgi, in 1998, triggered a wave of reviews on this organelle, including those focusing on Golgi architecture (Rambourg 1997; Farquhar and Palade 1998). More recent reviews that describe Golgi structure in great detail are provided by Marsh (2005) and Hua (2009). In this article, the most recent insights in mammalian Golgi architecture as revealed by distinct EM approaches are integrated into a general concept.  相似文献   

12.
Metabotropic glutamate receptors type 1 (mGluR1s) are required for a normal function of the mammalian brain. They are particularly important for synaptic signaling and plasticity in the cerebellum. Unlike ionotropic glutamate receptors that mediate rapid synaptic transmission, mGluR1s produce in cerebellar Purkinje cells a complex postsynaptic response consisting of two distinct signal components, namely a local dendritic calcium signal and a slow excitatory postsynaptic potential. The basic mechanisms underlying these synaptic responses were clarified in recent years. First, the work of several groups established that the dendritic calcium signal results from IP3 receptor-mediated calcium release from internal stores. Second, it was recently found that mGluR1-mediated slow excitatory postsynaptic potentials are mediated by the transient receptor potential channel TRPC3. This surprising finding established TRPC3 as a novel postsynaptic channel for glutamatergic synaptic transmission.Glutamate is the predominant neurotransmitter used by excitatory synapses in the mammalian brain (Hayashi 1952; Curtis et al. 1959). At postsynaptic sites, glutamate binds to two different classes of receptors, namely the ionotropic glutamate receptors (iGluRs) and the metabotropic glutamate receptors (mGluRs) (Sladeczek et al. 1985; Nicoletti et al. 1986; Sugiyama et al. 1987). The iGluRs represent ligand-gated nonselective cation channels that underlie excitatory postsynaptic currents (EPSCs). Based on their subunit composition, gating, and permeability properties, they are subdivided into three groups named after specific agonists: AMPA- (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid), NMDA receptors (N-methyl D-aspartate receptors) and kainate receptors (Alexander et al. 2009). The other class of glutamate receptors, the mGluRs, consists of receptors that are coupled to G proteins and act through distinct downstream signaling cascades. They are structurally different from iGluRs and characterized by the presence of seven transmembrane domains (Houamed et al. 1991; Masu et al. 1991). The mGluRs exist as homodimers that do not by themselves form an ion-permeable pore in the membrane (Ozawa et al. 1998). To date, eight different genes (and more splice variants) encoding mGluRs have been identified and form the mGluR1 through mGluR8 subtypes (Alexander et al. 2009). Based on the amino acid sequence homology, downstream signal transduction pathways, and pharmacological properties, each of the subtypes was assigned to one of three groups. Group I receptors consist of mGluR1 and mGluR5 that positively couple to the phospholipase C (PLC). The receptors mGluR2 and mGluR3 constitute group II, whereas the remaining mGluRs, namely mGluR4, mGluR6, mGluR7, and mGluR8, belong to group III. Both groups II and III inhibit the adenylyl cyclase and thereby reduce the concentration of cAMP in the cytosol.Of all different subtypes, mGluR1 is the most abundantly expressed mGluR in the mammalian central nervous system. In the brain, mGluR1 is highly expressed in the olfactory bulb, dentate gyrus, and cerebellum (Lein et al. 2007). The highest expression level of mGluR1 in the brain is found in Purkinje cells, the principal neurons of the cerebellar cortex (Shigemoto et al. 1992; Lein et al. 2007). Together with the AMPA receptors, mGluR1s are part of the excitatory synapses formed between parallel fibers and Purkinje cells (Fig. 1A). Each Purkinje cell is innervated by 100,000–200,000 parallel fibers (Ito 2006) that are axons of the cerebellar granule cells, the most abundant type of neuron in the brain. A second type of excitatory input to Purkinje cells is represented by the climbing fibers that originate in the inferior olive in the brain stem (Ito 2006). The two excitatory synaptic inputs to Purkinje cells are important determinants for the main functions of the cerebellum, including the real-time control of movement precision, error-correction, and control of posture as well as the procedural learning of complex movement sequences and conditioned responses.Open in a separate windowFigure 1.Parallel fiber-evoked mGluR1-dependent signals. (A) Diagram showing the parallel fiber synaptic input to Purkinje cell dendrites. (B) Microelectrode recording of glutamatergic postsynaptic potentials from a Purkinje cell in an acute slice of adult rat cerebellum. Short trains of stimuli to the parallel fibers (5–6 at 50 Hz) caused summation of the early AMPA receptor-dependent EPSPs (leading to spike firing) and a slow, delayed, depolarizing potential (slow EPSP), which was reversibly inhibited by antagonist of mGluRs (+)-MCPG (1mM). (C) Confocal image of a patch-clamped Purkinje cell in a cerebellar slice of an adult mouse. The patch-clamp pipette and the glass capillary used for electrical stimulation of parallel fibers are depicted schematically. The site of stimulation is shown at higher magnification in D. (D) Left: Parallel fiber-evoked (five pulses at 200 Hz, in 10 mM CNQX) synaptic responses consisting of a dendritic mGluR1-dependent Ca2+ transient (ΔF/F, top) and an early rapid and a slow excitatory postsynaptic current (EPSC, bottom). Block of the mGluR1-dependent components by the group I-specific mGluR-antagonist CPCCOEt (200 µM) is shown as indicated. Right: Pseudocolor image of the synaptic Ca2+ signal. (B, Reprinted with modifications, with permission, from Batchelor and Gaithwaite 1997 [Nature Publishing Group].)It is expected that mGluR1 is involved in many of these cerebellar functions. This view is supported by the observation that mGluR1-deficient knockout mice show severe impairments in motor coordination. In particular, the gait of these mice is strongly affected as well as their ability for motor learning and general coordination (Aiba et al. 1994). The phenotype of the general mGluR1-knockout mice is rescued by the insertion of the gene encoding mGluR1 exclusively into cerebellar Purkinje cells (Ichise et al. 2000) and blockade of mGluR1 expression only in Purkinje cells of adult mice leads to impaired motor coordination (Nakao et al. 2007). These findings established mGluR1 in Purkinje cell as synaptic receptors that are indispensable for a normal cerebellar function.Synaptic transmission involving mGluR1s is found at both parallel fiber-Purkinje cell synapses (Batchelor and Garthwaite 1993; Batchelor et al. 1994) as well as at climbing fiber-Purkinje cell synapses (Dzubay and Otis 2002). Most of our knowledge on the mGluR1 was gained from the analysis of the parallel fiber synapses. The parallel fiber synapse is quite unique in the central nervous system regarding its endowment with neurotransmitter receptors. In contrast to most other glutamatergic synapses in the mammalian brain, it lacks functional NMDA receptors (Shin and Linden 2005). The entire synaptic transmission at these synapses relies on AMPA receptors and on mGluR1 (Takechi et al. 1998). Although AMPA receptors are effectively activated even with single shock stimuli (Konnerth et al. 1990; Llano et al. 1991b), activation of mGluRs requires repetitive stimulation (Batchelor and Garthwaite 1993; Batchelor et al. 1994; Batchelor and Garthwaite 1997; Takechi et al. 1998). A possible explanation for the need of repetitive stimulation may relate to the observation that mGluR1s are found mostly at the periphery of the subsynaptic region (Nusser et al. 1994). At these sites outside the synaptic cleft, glutamate levels that are sufficiently high for receptor activation may be reached only with repetitive stimulation.At parallel fiber-Purkinje cell synapses, repetitive stimulation produces an initial AMPA receptor postsynaptic signal component, followed by a more prolonged mGluR1 component (Fig. 1). Figure 1B shows a current clamp recording of this response consisting of an early burst of action potentials, followed by a prolonged depolarization known as a “slow excitatory postsynaptic potential” (slow EPSP) (Batchelor and Garthwaite 1993; Batchelor et al. 1994; Batchelor and Garthwaite 1997). Voltage-clamp recordings allow a clear separation of the initial rapid, AMPA receptor mediated excitatory postsynaptic current (EPSC) and the mGluR1-mediated slow EPSC (Fig. 1D) (Takechi et al. 1998; Hartmann et al. 2008). In addition of inducing the slow EPSPs, mGluR1s mediate a large and highly localized dendritic calcium transient in cerebellar Purkinje cells (Fig. 1D) (Llano et al. 1991a; Finch and Augustine 1998; Takechi et al. 1998).  相似文献   

13.
The Desmosome     
Desmosomes are intercellular junctions that tether intermediate filaments to the plasma membrane. Desmogleins and desmocollins, members of the cadherin superfamily, mediate adhesion at desmosomes. Cytoplasmic components of the desmosome associate with the desmosomal cadherin tails through a series of protein interactions, which serve to recruit intermediate filaments to sites of desmosome assembly. These desmosomal plaque components include plakoglobin and the plakophilins, members of the armadillo gene family. Linkage to the cytoskeleton is mediated by the intermediate filament binding protein, desmoplakin, which associates with both plakoglobin and plakophilins. Although desmosomes are critical for maintaining stable cell–cell adhesion, emerging evidence indicates that they are also dynamic structures that contribute to cellular processes beyond that of cell adhesion. This article outlines the structure and function of the major desmosomal proteins, and explores the contributions of this protein complex to tissue architecture and morphogenesis.The desmosome is an adhesive intercellular junction that is crucial to tissues that experience mechanical stress, such as the myocardium, bladder, gastrointestinal mucosa, and skin (Getsios et al. 2004b; Holthofer et al. 2007). The desmosome was first observed in the spinous layer of epidermis by the Italian pathologist Giulio Bizzozero (1846–1901). Bizzozero''s observations of these small dense nodules, subsequently named “nodes of Bizzozero,” led him to the insightful interpretation of these structures as adhesive cell–cell contact points. The term desmosome was later coined by Josef Schaffer in 1920 and is derived from the Greek words “desmo,” meaning bond or fastening, and “soma,” meaning body (Wells 2005; Calkins and Setzer 2007). The introduction of electron microscopy yielded a series of advances by Porter, Odland, and Kelly in the 1950s and 1960s, which revealed desmosome organization at the ultrastructural level. These studies and others indicated that the desmosome can be divided into three morphologically identifiable zones: the extracellular core region (desmoglea), the outer dense plaque (ODP), and the inner dense plaque (IDP) (Fig. 1A) (Kowalczyk et al. 1994; Schmidt et al. 1994; Green and Jones 1996; North et al. 1999; Garrod and Chidgey 2008).Open in a separate windowFigure 1.A model for the structure of desmosomes. (A) Electron micrograph of a desmosome. (B) Schematic of desmosomal proteins and relative distance from the plasma membrane (PM). The desmosomal cadherins, the desmogleins and desmocollins, extend into extracellular core and outer dense plaque (ODP) to establish contact and adhere to neighboring cells in a Ca2+-dependent manner. The cadherin cytoplasmic tails associate linker proteins, plakoglobin (PG), the plakophilins (PKP), and desmoplakin (DP). DP binds to keratin intermediate filaments (KIF) within the inner dense plaque (IDP), serving to tether the intermediate filaments to the plasma membrane. (Adapted with permission from Kottke et al. 2006.)In the mid 1970s, Skerrow and Matoltsy (Skerrow and Matoltsy 1974a; Skerrow and Matoltsy 1974b) advanced the field by isolating desmosomes using biochemical approaches (Bass-Zubek and Green 2007).These landmark studies provided a foundation for the Franke and Steinberg laboratories to characterize the transmembrane glycoproteins and cytoplasmic plaque proteins that linked the structure to the intermediate filament cytoskeleton, and to develop immunological tools for localizing specific components (Franke et al. 1981; Kapprell et al. 1985; Steinberg et al. 1987). Collectively, these and other studies shaped our current view of how desmosomal components are organized.The transmembrane glycoproteins, termed desmogleins and desmocollins (Garrod and Chidgey 2008), represent separate subfamilies of the cadherin superfamily of calcium dependent adhesion molecules. The extracellular domains of the desmogleins and desmocollins mediate adhesion, whereas the cytoplasmic tails of these cadherins associate with the desmosomal plaque proteins. The outer dense plaque consists of the cytoplasmic tails of the desmosomal cadherins, which bind to members of the armadillo and plakin family of linker proteins (Kowalczyk et al. 1994; Getsios et al. 2004b; Garrod and Chidgey 2008). Plakoglobin, a member of the armadillo family, binds directly to the cytoplasmic tails of both the desmogleins and the desmocollins (Wahl et al. 1996; Witcher et al. 1996). Desmoplakin, a member of the plakin family, interacts with both plakoglobin and another subgroup of armadillo family proteins, the plakophilins (Cowin and Burke 1996). Finally, the interaction between desmoplakin and the keratin filaments forms the inner dense plaque, tethering the cytoskeletal network to the adhesion complex (Fig. 1B) (Kowalczyk et al. 1994; Getsios et al. 2004b; Garrod and Chidgey 2008).The following sections of this article describe the structural and functional characteristics of the major desmosomal proteins. In addition, we discuss differences in tissue expression patterns of desmosomal proteins and the role of desmosomes in human disease. A comprehensive review of additional proteins found to regulate or associate with desmosomes is provided elsewhere (Holthofer et al. 2007) and discussion of desmosome dynamics is provided in Green et al. 2009.  相似文献   

14.
The epidermal growth factor receptor (EGFR) was among the first receptor tyrosine kinases (RTKs) for which ligand binding was studied and for which the importance of ligand-induced dimerization was established. As a result, EGFR and its relatives have frequently been termed “prototypical” RTKs. Many years of mechanistic studies, however, have revealed that—far from being prototypical—the EGFR family is quite unique. As we discuss in this review, the EGFR family uses a distinctive “receptor-mediated” dimerization mechanism, with ligand binding inducing a dramatic conformational change that exposes a dimerization arm. Intracellular kinase domain regulation in this family is also unique, being driven by allosteric changes induced by asymmetric dimer formation rather than the more typical activation-loop phosphorylation. EGFR family members also distinguish themselves from other RTKs in having an intracellular juxtamembrane (JM) domain that activates (rather than autoinhibits) the receptor and a very large carboxy-terminal tail that contains autophosphorylation sites and serves an autoregulatory function. We discuss recent advances in mechanistic aspects of all of these components of EGFR family members, attempting to integrate them into a view of how RTKs in this important class are regulated at the cell surface.The epidermal growth factor receptor (EGFR) is often considered the “prototypical” receptor tyrosine kinase (RTK) and has been intensively studied. It is one of a family of four RTKs in humans, the others being ErbB2/HER2, ErbB3/HER3, and ErbB4/HER4 (Fig. 1). EGFR and its relatives are known oncogenic drivers in cancers such as lung cancer (Mok 2011), breast cancer (Arteaga et al. 2011), and glioblastoma (Libermann et al. 1985; Lee et al. 2006a; Vivanco et al. 2012), and inhibitors of these receptors have been among the most successful examples of targeted cancer therapies to date (Arteaga 2003; Moasser 2007; Zhang et al. 2007), including antibody therapeutics (e.g., trastuzumab and cetuximab) and small-molecule tyrosine kinase inhibitors (e.g., erlotinib, gefitinib, lapatinib).Open in a separate windowFigure 1.Schematic representation of EGFR/ErbB family receptors and their ligands. (A) The domain composition of human EGFR is shown. The extracellular region contains four domains: Domain I (amino acids 1–165), domain II (amino acids 165–310), domain III (amino acids 310–480), and domain IV (amino acids 480–620). Domains I and III are closely related in sequence, as are domains II and IV. Shown are representations of the structures of domains I and IV. Domain IV contains two types of disulfide-bonded module (C1 and C2). In C1 domains, a single disulfide constrains an intervening bow-like loop. In C2 modules, two disulfides link four successive cysteines in the patterns C1–C3 and C2–C4 to give a knot-like structure. A short extracellular juxtamembrane (eJM) region separates the extracellular region from the ∼23-amino-acid transmembrane (TM) domain. Within the cell, a short intracellular juxtamembrane (iJM) region separates the tyrosine kinase domain (TKD) from the membrane. A representative EGFR TKD structure is shown. The TKD is followed by a carboxy-terminal largely unstructured tail (amino acids 953–1186) that contains at least five tyrosine autophosphorylation sites. (B) EGFR is one of four members of the EGFR/ErbB family in humans. The other members are ErbB2/HER2, for which no soluble activating ligand is shown; ErbB3/HER3, which has a significantly impaired kinase domain (Jura et al. 2009b; Shi et al. 2010); and ErbB4/HER4. The primary active moiety of the ligands for these receptors is the EGF-like domain, shown as a cartoon structure (top right). EGFR is activated by the EGFR agonists: EGF itself, TGF-α (transforming growth factor α), ARG (amphiregulin), and EGN (epigen). The bispecific ligands regulate both EGFR and ErbB4: HB-EGF (heparin-binding EGF-like growth factor), EPR (epiregulin), and BTC (betacellulin). Neuregulins (NRGs) 1 and 2 regulate ErbB3 and ErbB4, whereas NRG3 and NRG4 appear to be specific for ErbB4 (Wilson et al. 2009).Far from being prototypical, however, it is now clear that regulation of EGFR family members is unique among RTKs (Ferguson 2008; Lemmon 2009; Lemmon and Schlessinger 2010). Structural studies have revealed how the ∼620-amino-acid isolated extracellular region is induced to dimerize after growth factor binding (Burgess et al. 2003) and how the isolated intracellular tyrosine kinase domain (TKD) becomes allosterically activated after forming an asymmetric dimer (Zhang et al. 2006; Jura et al. 2009a; Red Brewer et al. 2009). These findings have typically been interpreted in the context of a model in which EGF family receptors are regulated through ligand-induced receptor homodimerization or heterodimerization, with growth factor binding converting the receptor from an inactive monomeric configuration to an active dimeric conformation (Yarden and Schlessinger 1987; Schlessinger 1988, 2014; Ullrich and Schlessinger 1990). Although this original model has stood the test of time and initiated a whole field of studies of ligand-induced RTK dimerization, recent work has provided highly sophisticated views of a unique mode of allosteric regulation used by EGFR.  相似文献   

15.
The control of translation and mRNA degradation is important in the regulation of eukaryotic gene expression. In general, translation and steps in the major pathway of mRNA decay are in competition with each other. mRNAs that are not engaged in translation can aggregate into cytoplasmic mRNP granules referred to as processing bodies (P-bodies) and stress granules, which are related to mRNP particles that control translation in early development and neurons. Analyses of P-bodies and stress granules suggest a dynamic process, referred to as the mRNA Cycle, wherein mRNPs can move between polysomes, P-bodies and stress granules although the functional roles of mRNP assembly into higher order structures remain poorly understood. In this article, we review what is known about the coupling of translation and mRNA degradation, the properties of P-bodies and stress granules, and how assembly of mRNPs into larger structures might influence cellular function.The translation and decay of mRNAs play key roles in the control of eukaryotic gene expression. The determination of eukaryotic mRNA decay pathways has allowed insight into how translation and mRNA degradation are coupled. Degradation of eukaryotic mRNAs is generally initiated by shortening of the 3′ poly (A) tail (Fig. 1A) (reviewed in Parker and Song 2004; Garneau et al. 2007) by the major mRNA deadenylase, the Ccr4/Pop2/Not complex (Daugeron et al. 2001; Tucker et al. 2001; Thore et al. 2003). Following deadenylation, mRNAs can be degraded 3′ to 5′ by the exosome (Anderson and Parker 1998; Wang and Kiledjian 2001). However, more commonly, mRNAs are decapped by the Dcp1/Dcp2 decapping enzyme and then degraded 5′ to 3′ by the exonuclease, Xrn1 (Decker and Parker 1993; Hsu and Stevens 1993; Muhlrad et al. 1994, 1995; Dunckley and Parker 1999; van Dijk et al. 2002; Steiger et al. 2003). In metazoans, a second decapping enzyme, Nudt16, also contributes to mRNA turnover (Song et al. 2010).Open in a separate windowFigure 1.Eukaryotic mRNA decay pathways. (A) General mRNA decay pathways. (B) Specialized decay pathways that degrade translationally aberrant mRNAs.The processes of mRNA decay and translation are interconnected in eukaryotic cells in many ways. For example, quality control mechanisms exist to detect aberrancies in translation, which then lead to mRNAs being degraded by specialized mRNA decay pathways (Fig. 1B). Nonsense-mediated decay (NMD) is one such mRNA quality control system that degrades mRNAs that terminate translation aberrantly. In yeast, aberrant translation termination leads to deadenylation-independent decapping (Muhlrad and Parker 1994), whereas in metazoan cells NMD substrates can be both decapped and endonucleolytically cleaved and degraded (reviewed in Isken and Maquat 2007). A second quality control system for mRNA translation is referred to as no-go decay (NGD) and leads to endonucleolytic cleavage of mRNAs with strong stalls in translation elongation (Doma and Parker 2006; reviewed in Harigaya and Parker 2010). Another mechanism of mRNA quality control is the rapid 3′ to 5′ degradation of mRNAs that do not contain translation termination codons, which is referred to as non-stop decay (NSD) (Frischmeyer et al. 2002; van Hoof et al. 2002). The available evidence suggests these specialized mechanisms function primarily on aberrant mRNAs that are produced by defects in splicing, 3′ end formation, or damage to RNAs.The main pathway of mRNA degradation is also in competition with translation initiation. Competition between the two processes was first suggested by the observation that removal of the poly (A) tail and the cap structure, both of which stimulate translation initiation, were the key steps in mRNA degradation. In addition, inhibition of translation initiation by strong secondary structures in the 5′UTR, translation initiation inhibitors, a poor AUG context, or mutations in initiation factors increases the rates of deadenylation and decapping (Muhlrad et al. 1995; Muckenthaler et al. 1997; Lagrandeur and Parker 1999; Schwartz and Parker 1999). Moreover, the cap binding protein eIF4E, known to stimulate translation initiation, inhibits the decapping enzyme, Dcp1/Dcp2, both in vivo and in vitro (Schwartz and Parker 1999; Schwartz and Parker 2000). Finally, many mRNA specific regulatory factors, (e.g., miRNAs or PUF proteins), both repress translation and accelerate deadenylation and decapping (reviewed in Wickens et al. 2002; Behm-Ansmant et al. 2006; Franks and Lykke-Anderson 2008; Shyu et al. 2008).In the simplest model, the competition between translation and mRNA degradation can be understood through changes in the proteins bound to the cap and poly (A) tail that then influence the accessibility of these structures to deadenylases and decapping enzymes. For example, given that the Ccr4/Pop2/Not deadenylase complex is inhibited by poly (A)-binding protein (Pab1) (Tucker et al. 2002), the effects of translation on deadenylation are most likely through dynamic changes in the association of Pab1 binding with the poly (A) tail. One possibility is that defects in translation initiation either directly or indirectly decrease Pab1 association with the poly (A) tail. Deadenylation is also affected by aspects of translation termination. For instance, premature translation termination in yeast accelerates poly (A) shortening as part of the process of NMD (Cao and Parker 2003; Mitchell and Tollervey 2003). The coupling of translation termination to deadenylation has been suggested to occur through direct interactions of the translation termination factor eRF3 with Pab1 (Cosson et al. 2002), which may lead to Pab1 transiently dissociating from the poly (A) tail. Interestingly, in yeast, once the poly (A) tail reaches an oligo (A) length of 10–12 residues, a length that reduces the affinity of Pab1, the mRNA can become a substrate for decapping and for binding of the Pat1/Lsm1-7 complex (Tharun and Parker 2001; Chowdhury et al. 2007), which enhances the rate of decapping. This exchange of the Pab1 protein for the Pat1/Lsm1-7 complex is part of the mechanism that allows decapping to be promoted following deadenylation.A similar mRNP dynamic is also likely to occur on the cap structure. Specifically, the competition between translation initiation and decapping suggests that prior to decapping, translation initiation factors are exchanged for decapping factors, thereby assembling a distinct “decapping” mRNP that is no longer capable of translation initiation (Tharun and Parker 2001). This idea is supported by the observation that some decapping activators also function as translational repressors (Coller and Parker 2005; Pilkington and Parker 2008; Nissan et al. 2010). Thus, mRNA decapping appears to occur in two steps, first inhibition of translation initiation and exchange of translation factors for the general repression/degradation machinery, and a second step whereby the mRNA is actually degraded. Thus, by understanding the changes in mRNP states between actively translating mRNAs and mRNAs that are translationally repressed and possibly stored or ultimately degraded we will better understand how the fate of mRNAs is controlled in the cytoplasm.  相似文献   

16.
Receptor tyrosine kinases (RTKs) and their cellular signaling pathways play important roles in normal development and homeostasis. Aberrations in their activation or signaling leads to many pathologies, especially cancers, motivating the development of a variety of drugs that block RTK signaling that have been successfully applied for the treatment of many cancers. As the current field of RTKs and their signaling pathways are covered by a very large amount of literature, spread over half a century, I am focusing the scope of this review on seminal discoveries made before tyrosine phosphorylation was discovered, and on the early days of research into RTKs and their cellular signaling pathways. I review the history of the early days of research in the field of RTKs. I emphasize key early findings, which provided conceptual frameworks for addressing the questions of how RTKs are activated and how they regulate intracellular signaling pathways.The family of cell-surface receptors designated receptor tyrosine kinases (RTK) received their name more that a decade after the same molecules were already known as the cell-surface receptors for insulin (insulin receptor), epidermal growth factor (EGFR), and many other growth factor receptors. Following the pioneering discoveries of nerve growth factor and epidermal growth factor (EGF; Levi-Montalcini and Booker 1960; Cohen 1962) and the establishment of the important roles of these two growth factors in the control of neuronal differentiation and cell proliferation in vivo and in vitro, it became clear that these cytokines bind specifically to cell-surface receptors. Insulin had already been discovered by this time, and had been applied successfully to treat diabetes patients since the early twentieth century. The resulting homogenous preparations of pure insulin enabled the quantitative characterization of insulin binding to its receptor on intact cells or to solubilized insulin receptor preparations using radiolabeled insulin (De Meyts et al. 1973). These studies greatly advanced understanding of the ligand binding characteristics of insulin receptor and, later on EGFR (Carpenter et al. 1975), including the establishment of negative cooperativity in insulin binding to its receptor expressed on the surface of living cells (De Meyts et al. 1973). Moreover, these studies shed important light on the dynamic nature of the cellular behavior of these receptors. The capacities of insulin receptor and EGFR to undergo ligand-dependent down-regulation and desensitization through receptor-mediated internalization and degradation (Carpenter and Cohen 1976; Gordon et al. 1978; Schlessinger et al. 1978a,b; Carpentier et al. 1979; Haigler et al. 1979) were also established well before the realization that growth factors receptors are endowed with intrinsic protein tyrosine kinase activities (Fig. 1).Open in a separate windowFigure 1.A time line of key findings during the history of RTKs, with emphasis on findings and discoveries that produced the conceptual framework in the development of the RTK field and its application for cancer therapy. References for the key findings are also presented in the text (Lee et al. 1985; Libermann et al. 1985; Margolis et al. 1990; Bottaro et al. 1991; Bae et al. 2009).Progress was also made in elucidating the role of growth factors in normal embryonic development, wound healing, and pathological conditions such as cancer. Early studies in the 1960s and 1970s showed that growth factors play an important role in oncogenesis induced by retroviruses and in the proliferation of tumor-derived cancer cells. Pioneering studies performed by Howard Temin (1966, 1967) showed that cancer cells need less insulin and serum growth factors for cell proliferation compared with normal cells, suggesting that cancer cells produce and use their own growth factors and/or use cellular processes that in normal cells are regulated by exogenously supplied growth factors; both predictions were subsequently confirmed. A variety of new polypeptide growth factors that stimulate cell proliferation by binding to receptors at the cell surface were subsequently discovered. Those include a growth factor isolated from human platelets designated platelet-derived growth factor (PDGF; Antoniades et al. 1979; Heldin et al. 1979), a growth factor isolated from bovine brain designated fibroblast growth factor (FGF; Gospodarowicz et al. 1978), a growth factor isolated from rat platelets that stimulates the proliferation of mature hepatocytes, designated hepatocyte growth factor (HGF; Nakamura et al. 1986). In addition to EGF, another growth factor that binds selectively to cells expressing EGFR was isolated from virally and chemically transformed cells, suggesting that this growth factor—designated transforming growth factor α—may play a role in oncogenesis by an autocrine mechanism (Roberts et al. 1980, 1982). This discovery provided further support to the earlier finding that transformation by murine and feline sarcoma viruses selectively interferes with EGF binding to EGFR in transformed cells (Todaro et al. 1976). Together with many other studies published since the 1980s, this work showed that growth factors and their receptors play numerous important roles during development and in many normal cellular processes as well as in pathologies such as cancer, diabetes, atherosclerosis, severe bone disorders, and tumor angiogenesis.Visualization of dynamic cellular redistribution of ligand/receptor complexes, and rapid receptor-mediated internalization of growth factors such as insulin or EGF, led to the proposal that cell-surface receptors for these ligands may play a passive role in delivering them to intracellular compartments in which internalized EGF or insulin molecules exert their actions (Vigneri et al. 1978; Podlecki et al. 1986; Jiang and Schindler 1990). In other words, according to this hypothesis, the biological signals induced by insulin or EGF were thought to be mediated by binding of the ligands themselves to intracellular target(s) in the cytoplasm or nucleus, with the role of the cell-surface receptor being to act as a “carrier” that delivers them directly to these targets. An alternative hypothesis was that insulin or EGF activates their cognate receptors at the cell surface, which in turn stimulate the production of an intracellular second messenger molecule analogous to cAMP in signaling by the G-protein-activating β-adrenergic receptor. Indeed, several potential second messengers that are generated in cells on stimulation with insulin or other growth factors were proposed before (and even after) it became clear that insulin receptor, EGFR, and other RTKs are endowed with intrinsic tyrosine kinase activity (Larner et al. 1979; Das 1980; Saltiel and Cuatrecasas 1986).A demonstration that anti-insulin receptor antibodies from the serum of certain diabetic patients could mimic cellular responses of insulin (Flier et al. 1977; Van Obberghen et al. 1979) provided the first conclusive answer to the question of whether the biological activity of growth factors is mediated directly or indirectly through their membrane receptors. This experiment ruled out the possibility that insulin receptor functions as a passive carrier that delivers insulin to an intracellular target to induce cellular responses. Studies showing that intact, bivalent antibodies against the insulin receptor can activate its signaling, whereas monovalent Fab fragments of the same antibodies cannot further argued that ligand-induced receptor dimerization or stimulation of a particular arrangement between two receptor molecules in a dimer can activate the insulin receptor (Kahn et al. 1978).A similar conclusion was reached using certain monoclonal antibodies that bind to the extracellular region of EGFR and block ligand binding (Schreiber et al. 1981). Whereas intact antibodies were able to mimic EGF in stimulating a variety of EGF-like responses including cell proliferation, monovalent Fab fragments of the same monoclonal EGFR antibodies failed to do so—and acted instead as EGFR antagonists (Schreiber et al. 1981, 1983). These experiments provided strong evidence both that EGFR plays a crucial role in mediating EGF-induced cellular responses and that EGFR is activated by ligand-induced receptor dimerization (Schreiber 1981, 1983).  相似文献   

17.
Vascular endothelial growth factor receptors (VEGFRs) in vertebrates play essential roles in the regulation of angiogenesis and lymphangiogenesis. VEGFRs belong to the receptor-type tyrosine kinase (RTK) supergene family. They consist of a ligand-binding region with seven immunoglobulin (7 Ig) -like domains, a trans-membrane (TM) domain, and a tyrosine kinase (TK) domain with a long kinase insert (KI) (also known as a type-V RTK). Structurally, VEGFRs are distantly related to the members of the M-colony stimulating factor receptor/platelet-derived growth factor receptor (CSFR)/(PDGFR) family, which have five immunoglobulin (5 Ig)-like domains. However, signal transduction in VEGFRs significantly differs from that in M-CSFR/PDGFRs. VEGFR2, the major signal transducer for angiogenesis, preferentially uses the phospholipase Cγ-protein kinase C (PLC-γ-PKC)-MAPK pathway, whereas M-CSFR/PDGFRs use the PI3 kinase-Ras-MAPK pathway for cell proliferation. In phylogenetic development, the VEGFR-like receptor in nonvertebrates appears to be the ancestor of the 7 Ig- and 5 Ig-RTK families because most nonvertebrates have only a single 7 Ig-RTK gene. In mammals, VEGFRs are deeply involved in pathological angiogenesis, including cancer and inflammation. Thus, an efficient inhibitor targeting VEGFRs could be useful in suppressing various diseases.Angiogenesis, blood vessel formation, is essential for supplying oxygen and nutrients to the tissues and for removing carbon dioxide and waste materials (Hanahan and Folkman 1996; Risau 1997). Furthermore, lymph vessels are crucial for the absorption of tissue fluids and the recovery of tissue-infiltrated lymphocytes. In the 1980s, two proteins, vascular permeability factor (VPF) and vascular endothelial growth factor receptors (VEGF), were independently isolated. However, in 1989, these proteins were found to be identical and encoded by a single gene (Ferrara and Davis-Smith 1997; Dvorak 2002). This protein, named VEGF (or VEGF-A), has a homodimeric structure with three intramolecular and two intermolecular disulfide (S–S) bonds. However, its receptor and signaling patterns were yet to be elucidated.In 1990, we isolated from human placenta a novel receptor-type tyrosine kinase (RTK) cDNA encoding a protein with seven Ig-like domains in its extracellular region. We designated it Flt-1 (Fms-like tyrosine kinase-1; the first type-V RTK) because of its similarity with the Fms (M-CSFR) and PDGFR family (Fig. 1) (Shibuya et al. 1990; Shibuya 1995). On the basis of this similarity, the ligand of Flt-1 was suggested to be a protein with a homodimeric structure, such as M-CSF and PDGF. In 1992, De Vries et al. (1992) reported tight binding and activation of Flt-1 with VEGF, indicating that Flt-1 is the first receptor for VEGF (VEGFR1).Open in a separate windowFigure 1.The VEGF-VEGFR system and its inhibitors. VEGF (VEGF-A) and its receptors, VEGFR1 and VEGFR2, play a major role in vasculogenesis and angiogenesis. VEGFR3 regulates not only lymphangiogenesis but also angiogenesis under pathological conditions. Neuropilins (Nrp)-1 and Nrp-2 function as coreceptors for VEGFR1 and VEGFR2, respectively, and efficiently stimulate VEGFR signaling. A heterodimer formed between two receptors, such as VEGFR1/VEGFR2 and VEGFR2/VEGFR3, was reported to regulate angiogenesis as well as lymphangiogenesis. Various inhibitors were developed to suppress this system, and some of them are now widely used clinically.Several years later, two RTKs structurally related to Flt-1 were isolated: one was KDR/Flk-1 (kinase-insert [KI] domain receptor in humans/fetal liver kinase-1 in mice; VEGFR2), while the other was Flt-4 (VEGFR3) (Matthews et al. 1991; Terman et al. 1991; Alitalo and Carmeliet 2002). VEGF binds VEGFR1 and VEGFR2, but not VEGFR3. Other VEGF family members, VEGF-C and VEGF-D, bind VEGFR3 and activate it for lymphangiogenesis (Joukov et al. 1997; Achen et al. 1998).When we isolated full-length flt-1 cDNA, we also found a short 3-kb-long flt-1 mRNA, encoding a peptide without a TK domain that was highly expressed in normal placenta (Shibuya et al. 1990). This short flt-1 mRNA was later reported to encode a soluble Flt-1 (sFlt-1) protein without trans-membrane (TM) and TK domains (Kendall and Thomas 1993; Hornig et al. 2000; Helske et al. 2001). Soluble Flt-1 is strongly expressed in the placental trophoblasts and is abnormally expressed in preeclampsia (Koga et al. 2003; Maynard et al. 2003). The main function of sFlt-1 is considered to be trapping of endogenous VEGF. Thus, extreme trapping and suppression of VEGF in various tissues of the body may result in the hypertension and proteinuria observed in preeclampsia patients (Levine et al. 2004).VEGF heterozygotic knockout mice (VEGF-A+/− mice) are embryonic lethal because of immature development of angiogenesis, indicating that the concentration of VEGF is tightly regulated in tissues during embryogenesis (Carmeliet et al. 1996; Ferrara et al. 1996). The VEGF-VEGFR system is the major regulator of angiogenesis. Therefore, this system is an attractive target for antiangiogenic therapy and proangiogenic therapy.  相似文献   

18.
The nuclear factor κB (NF-κB) pathways play a major role in Drosophila host defense. Two recognition and signaling cascades control this immune response. The Toll pathway is activated by Gram-positive bacteria and by fungi, whereas the immune deficiency (Imd) pathway responds to Gram-negative bacterial infection. The basic mechanisms of recognition of these various types of microbial infections by the adult fly are now globally understood. Even though some elements are missing in the intracellular pathways, numerous proteins and interactions have been identified. In this article, we present a general picture of the immune functions of NF-κB in Drosophila with all the partners involved in recognition and in the signaling cascades.The paramount roles of NF-κB family members in Drosophila development and host defense are now relatively well established and have been the subject of several in-depth reviews in recent years, including some from this laboratory (e.g., Hoffmann 2003; Minakhina and Steward 2006; Ferrandon et al. 2007; Lemaitre and Hoffmann 2007; Aggarwal and Silverman 2008). To avoid excessive duplication, we limit this text to the general picture that has evolved over nearly two decades—since the initial demonstration that the dorsal gene plays a role in dorsoventral patterning in embryogenesis of Drosophila and that it encodes a member of the NF-κB family of inducible transactivators (Nüsslein-Volhard et al. 1980; Steward 1987; Roth et al. 1989). In the early nineties, it became apparent that NF-κB also plays a role in the antimicrobial host defense of Drosophila (Engström et al. 1993; Ip et al. 1993; Kappler et al. 1993; Reichhart et al. 1993). We focus in this article on the immune functions of NF-κB and refer the reader to recent reviews for the roles of NF-κB in development (Roth 2003; Brennan and Anderson 2004; Moussian and Roth 2005; Minakhina and Steward 2006).The Drosophila genome codes for three NF-κB family members (Fig. 1). Dorsal and DIF (for dorsal-related immunity factor) are 70 kDa proteins, with a typical Rel homology domain, which is 45% identical to that of the mammalian counterparts c-Rel, Rel A, and Rel B. Dorsal and DIF lie some 10 kbp apart on the second chromosome and probably arose from a recent duplication (Meng et al. 1999). Both proteins are retained in the cytoplasm by binding to the same 54-kDa inhibitor protein Cactus, which is homologous to mammalian IκBs (Schüpbach and Wieshaus 1989; Geisler et al. 1992). The single Drosophila Cactus gene is closest to mammalian IκBα (Huguet et al. 1997). The third member of the family in Drosophila, Relish, is a 100-kDa protein with an amino-terminal Rel domain and a carboxy-terminal extension with typical ankyrin repeats, as found in Cactus and mammalian IκBs. Relish is similar to mammalian p100 and p105 and its activation requires proteolytic cleavage as in the case for these mammalian counterparts (reviewed in Hultmark 2003).Open in a separate windowFigure 1.The NF-κB and IκB proteins in Drosophila. The length in amino acids is indicated by numbers. REL, Rel-homology domain; NLS, nuclear localization sequence; PEST, proline, glutamic acid, serine, and threonine-rich segment; Ac, acidic domain.Put in simple terms, NF-κB family members function in the host defense of Drosophila to control the expression of genes encoding immune-responsive peptides and proteins. Prominent among the induced genes are those encoding peptides with direct antimicrobial activity. To exert this function, Dorsal and DIF are translocated to the nucleus following stimulus-induced degradation of the inhibitor Cactus, whereas Relish requires stimulus-induced proteolytic cleavage for nuclear translocation of its amino-terminal Rel domain. This paradigm is similar to that observed in mammalian immunity. Again, for the sake of simplicity, we may say that the stimulus-induced degradation of Cactus, and the concomitant release of Dorsal or DIF, is primarily observed during Gram-positive bacterial and fungal infections and mediated by the Toll signaling pathway. In contrast, stimulus-induced proteolytic cleavage of Relish, and concomitant nuclear translocation of its amino-terminal Rel domain, is the hallmark of the response to Gram-negative bacterial infection and mediated by the Imd signaling pathway. Whether these pathways are also involved in the multifaceted defense against viruses remains an open question (Zambon et al. 2005). The Toll pathway was further shown to be involved in hematopoiesis of flies (Qiu et al. 1998). Of note, the Cactus-NF-κB module also plays a central role in the elimination of Plasmodium parasites in infected mosquitoes (Frolet et al. 2006). In the following, we review our information of the two established signaling pathways, Toll and Imd, which lead to gene reprogramming through NF-κB in response to bacterial and fungal infections. We first consider the upstream mechanisms that mediate the recognition of infection and allow for a certain level of discrimination between invading microorganisms. Gene reprogramming in this context is best illustrated by the induction of the antimicrobial peptide genes, which serve as the most convenient readouts of the antimicrobial defense of Drosophila (see Samakovlis et al. 1990; Reichhart et al. 1992; Ferrandon et al. 1998). Flies produce at least seven families of mostly cationic, small-sized, membrane-active peptides, with spectra variously directed against Gram-positive (defensins) and Gram-negative (diptericins, attacins, and drosocin) bacteria, and against fungi (drosomycins and metchnikowins), or with overlapping spectra (cecropins) (reviewed in Bulet et al. 1999; Hetru et al. 2003). The primary site of biosynthesis of these peptides is the fat body, a functional equivalent of the mammalian liver. Blood cells also participate in the production of antimicrobial peptides. As a rule, these molecules are secreted into the hemolymph where they reach remarkably high concentrations to oppose invading microorganisms (Hetru et al. 2003). This facet of the antimicrobial host defense is generally referred to as systemic immune response. Of note, the gut and the tracheae also produce antimicrobial peptides in response to microbes (see Tzou et al. 2000; Onfelt Tingvall et al. 2001; Liehl et al. 2006; Nehme et al. 2007).During infection, the Toll and Imd pathways control the expression of hundreds of genes. In addition to the antimicrobial peptides, these genes encode proteases, putative cytokines, cytoskeletal proteins, and many peptides and proteins whose function in the host defense are still not understood (De Gregorio et al. 2001; Irving et al. 2001).  相似文献   

19.
Fibroblast growth factors (FGFs) signal in a paracrine or endocrine fashion to mediate a myriad of biological activities, ranging from issuing developmental cues, maintaining tissue homeostasis, and regulating metabolic processes. FGFs carry out their diverse functions by binding and dimerizing FGF receptors (FGFRs) in a heparan sulfate (HS) cofactor- or Klotho coreceptor-assisted manner. The accumulated wealth of structural and biophysical data in the past decade has transformed our understanding of the mechanism of FGF signaling in human health and development, and has provided novel concepts in receptor tyrosine kinase (RTK) signaling. Among these contributions are the elucidation of HS-assisted receptor dimerization, delineation of the molecular determinants of ligand–receptor specificity, tyrosine kinase regulation, receptor cis-autoinhibition, and tyrosine trans-autophosphorylation. These structural studies have also revealed how disease-associated mutations highjack the physiological mechanisms of FGFR regulation to contribute to human diseases. In this paper, we will discuss the structurally and biophysically derived mechanisms of FGF signaling, and how the insights gained may guide the development of therapies for treatment of a diverse array of human diseases.Fibroblast growth factor (FGF) signaling fulfills essential roles in metazoan development and metabolism. A wealth of literature has documented the requirement for FGF signaling in multiple processes during embryogenesis, including implantation (Feldman et al. 1995), gastrulation (Sun et al. 1999), somitogenesis (Dubrulle and Pourquie 2004; Wahl et al. 2007; Lee et al. 2009; Naiche et al. 2011; Niwa et al. 2011), body plan formation (Martin 1998; Rodriguez Esteban et al. 1999; Tanaka et al. 2005; Mariani et al. 2008), morphogenesis (Metzger et al. 2008; Makarenkova et al. 2009), and organogenesis (Goldfarb 1996; Kato and Sekine 1999; Sekine et al. 1999; Sun et al. 1999; Colvin et al. 2001; Serls et al. 2005; Vega-Hernandez et al. 2011). Recent clinical and biochemical data have uncovered unexpected roles for FGF signaling in metabolic processes, including phosphate/vitamin D homeostasis (Consortium 2000; Razzaque and Lanske 2007; Nakatani et al. 2009; Gattineni et al. 2011; Kir et al. 2011), cholesterol/bile acid homeostasis (Yu et al. 2000a; Holt et al. 2003), and glucose/lipid metabolism (Fu et al. 2004; Moyers et al. 2007). Highlighting its diverse biology, deranged FGF signaling contributes to many human diseases, such as congenital craniosynostosis and dwarfism syndromes (Naski et al. 1996; Wilkie et al. 2002, 2005), Kallmann syndrome (Dode et al. 2003; Pitteloud et al. 2006a), hearing loss (Tekin et al. 2007, 2008), and renal phosphate wasting disorders (Shimada et al. 2001; White et al. 2001), as well as many acquired forms of cancers (Rand et al. 2005; Pollock et al. 2007; Gartside et al. 2009; di Martino et al. 2012). Endocrine FGFs have also been implicated in the progression of acquired metabolic disorders, including chronic kidney disease (Fliser et al. 2007), obesity (Inagaki et al. 2007; Moyers et al. 2007; Reinehr et al. 2012), and insulin resistance (Fu et al. 2004; Chen et al. 2008b; Chateau et al. 2010; Huang et al. 2011), giving rise to many opportunities for drug discovery in the field of FGF biology (Beenken and Mohammadi 2012).Based on sequence homology and phylogeny, the 18 mammalian FGFs are grouped into six subfamilies (Ornitz and Itoh 2001; Popovici et al. 2005; Itoh and Ornitz 2011). Five of these subfamilies act in a paracrine fashion, namely, the FGF1 subfamily (FGF1 and FGF2), the FGF4 subfamily (FGF4, FGF5, and FGF6), the FGF7 subfamily (FGF3, FGF7, FGF10, and FGF22), the FGF8 subfamily (FGF8, FGF17, and FGF18), and the FGF9 subfamily (FGF9, FGF16, and FGF20). In contrast, the FGF19 subfamily (FGF19, FGF21, and FGF23) signals in an endocrine manner (Beenken and Mohammadi 2012). FGFs exert their pleiotropic effects by binding and activating the FGF receptor (FGFR) subfamily of receptor tyrosine kinases that are coded by four genes (FGFR1, FGFR2, FGFR3, and FGFR4) in mammals (Johnson and Williams 1993; Mohammadi et al. 2005b). The extracellular domain of FGFRs consists of three immunoglobulin (Ig)-like domains (D1, D2, and D3), and the intracellular domain harbors the conserved tyrosine kinase domain flanked by the flexible amino-terminal juxtamembrane linker and carboxy-terminal tail (Lee et al. 1989; Dionne et al. 1991; Givol and Yayon 1992). A unique feature of FGFRs is the presence of a contiguous segment of glutamic and aspartic acids in the D1–D2 linker, termed the acid box (AB). The two-membrane proximal D2 and D3 and the intervening D2–D3 linker are necessary and sufficient for ligand binding/specificity (Dionne et al. 1990; Johnson et al. 1990), whereas D1 and the D1–D2 linker are implicated in receptor autoinhibition (Wang et al. 1995; Roghani and Moscatelli 2007; Kalinina et al. 2012). Alternative splicing and translational initiation further diversify both ligands and receptors. The amino-terminal regions of FGF8 and FGF17 can be differentially spliced to yield FGF8a, FGF8b, FGF8e, FGF8f (Gemel et al. 1996; Blunt et al. 1997), and FGF17a and FGF17b isoforms (Xu et al. 1999), whereas cytosine-thymine-guanine (CTG)-mediated translational initiation gives rise to multiple high molecular weight isoforms of FGF2 and FGF3 (Florkiewicz and Sommer 1989; Prats et al. 1989; Acland et al. 1990). The tissue-specific alternative splicing in D3 of FGFR1, FGFR2, and FGFR3 yields “b” and “c” receptor isoforms which, along with their temporal and spatial expression patterns, is the major regulator of FGF–FGFR specificity/promiscuity (Orr-Urtreger et al. 1993; Ornitz et al. 1996; Zhang et al. 2006). A large body of structural data on FGF–FGFR complexes has begun to reveal the intricate mechanisms by which different FGFs and FGFRs combine selectively to generate quantitatively and qualitatively different intracellular signals, culminating in distinct biological responses. In addition, these structural data have unveiled how pathogenic mutations hijack the normal physiological mechanisms of FGFR regulation to lead to pathogenesis. We will discuss the current state of the structural biology of the FGF–FGFR system, lessons learned from studying the mechanism of action of pathogenic mutations, and how the structural data are beginning to shape and advance the translational research.  相似文献   

20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号