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 31 毫秒
1.
The extracellular matrix (ECM) serves diverse functions and is a major component of the cellular microenvironment. The ECM is a highly dynamic structure, constantly undergoing a remodeling process where ECM components are deposited, degraded, or otherwise modified. ECM dynamics are indispensible during restructuring of tissue architecture. ECM remodeling is an important mechanism whereby cell differentiation can be regulated, including processes such as the establishment and maintenance of stem cell niches, branching morphogenesis, angiogenesis, bone remodeling, and wound repair. In contrast, abnormal ECM dynamics lead to deregulated cell proliferation and invasion, failure of cell death, and loss of cell differentiation, resulting in congenital defects and pathological processes including tissue fibrosis and cancer. Understanding the mechanisms of ECM remodeling and its regulation, therefore, is essential for developing new therapeutic interventions for diseases and novel strategies for tissue engineering and regenerative medicine.The extracellular matrix (ECM) forms a milieu surrounding cells that reciprocally influences cellular function to modulate diverse fundamental aspects of cell biology (Hynes 2009). The diversity and sophistication of ECM components and their respective cell surface receptors are among the most salient features during metazoan evolution (Har-el and Tanzer 1993; Hutter et al. 2000; Whittaker et al. 2006; Engler et al. 2009; Huxley-Jones et al. 2009; Ozbek et al. 2010). The ECM is extremely versatile and performs many functions in addition to its structural role. As a major component of the microenvironment of a cell, the ECM takes part in most basic cell behaviors, from cell proliferation, adhesion and migration, to cell differentiation and cell death (Hynes 2009). This pleiotropic aspect of ECM function depends on the highly dynamic structure of ECM and its remodeling as an effective mechanism whereby diverse cellular behaviors can be regulated. This concept is particularly important when considering processes and cell behaviors that need to be deployed promptly and transiently and wherein cell–cell and cell–matrix interactions are constantly changing (Daley et al. 2008).ECM dynamics are a feature of tissues wherein radical remodeling occurs, such as during metamorphosis of insects and amphibians or remodeling of the adult bone and mammary gland, and in developmental processes, including neural crest migration, angiogenesis, tooth and skeletal development, branching morphogenesis, maturation of synapses, and the nervous system (Berardi et al. 2004; Fukumoto and Yamada 2005; Page-McCaw et al. 2007; Zimmermann and Dours-Zimmermann 2008).ECM dynamics can result from changes of ECM composition, for example, because of altered synthesis or degradation of one or more ECM components, or in architecture because of altered organization. Mounting evidence has shown how individual ECM components are laid down, cross-linked, and organized together via covalent and noncovalent modifications and how they can greatly influence the fundamental aspects of cell behavior (Lopez et al. 2008; Engler et al. 2009; Egeblad et al. 2010b). This higher level of ECM organization is also dynamic and subject to sustained remodeling as mediated by reciprocal interactions between the ECM and its resident cellular components (Daley et al. 2008). Understandably, ECM dynamics are tightly regulated to ensure normal development, physiology, and robustness of organ systems. This is achieved by redundant mechanisms to modulate the expression and function of ECM modifying enzymes at multiple levels. When such control mechanisms are corrupted, ECM dynamics become deregulated, leading to various human congenital defects and diseases, including cancer.Here, we examine the players involved in ECM remodeling and how they are tightly regulated to achieve a delicate balance between stability and remodeling of the ECM. We focus on the cellular and molecular mechanisms through which ECM dynamics influence cellular behaviors. We illustrate how a wide variety of cell behaviors can be deployed by exploiting the important roles of ECM dynamics to build vertebrate organs and maintain their functions, and how deregulation of ECM dynamics contributes to the initiation and progression of human cancer.  相似文献   

2.
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   

3.
4.
Cell adhesions mediate important bidirectional interactions between cells and the extracellular matrix. They provide an interactive interface between the extracellular chemical and physical environment and the cellular scaffolding and signaling machinery. This dynamic, reciprocal regulation of intracellular processes and the matrix is mediated by membrane receptors such as the integrins, as well as many other components that comprise the adhesome. Adhesome constituents assemble themselves into different types of cell adhesion structures that vary in molecular complexity and change over time. These cell adhesions play crucial roles in cell migration, proliferation, and determination of cell fate.With the emergence of metazoan life approximately 600 million years ago, new biological mechanisms arose during the evolution of multicellular organisms with a defined body plan. These mechanisms of cell adhesion are a fundamental feature of all metazoans, from sponges to humans; they enable cells to attach to each other or to an extracellular matrix (ECM), cementing them together and organizing them into a coherent whole. The formation of adhesions and the regulation of their dynamics are crucial for embryogenesis, immune cell function, and wound repair, but they also contribute to disease, including cancer invasion and metastasis, or immune disorders (Hay 1991; Hynes 2002; Berrier and Yamada 2007; Alberts et al. 2008; Mory et al. 2008; Dubash et al. 2009; Manevich-Mendelson et al. 2009; Svensson et al. 2009; Wolfenson et al. 2009a). Adhesive interactions can occur with remarkable temporal and spatial precision. As illustrated in Figure 1, they not only link cells together into functional tissues and organs, but they also convey to the adhering cells accurate positional information concerning their cellular and extracellular environment. This information can, in turn, affect all facets of the cell’s life—its proliferation, differentiation, and fate. In addition to responding to the matrix, cell adhesions can actively remodel and restructure the ECM, driving a reciprocal, bidirectional interaction between the cell and its surrounding matrix. These two fundamental aspects of cell–ECM adhesion—physical/structural roles and environmental sensing/signaling, as well as the dynamic molecular interrelationships between them—will be the primary subjects of this article.Open in a separate windowFigure 1.Schematic illustration highlighting the dynamic cross talk between cells and the extracellular matrix (ECM). Cells secrete and remodel the ECM, and the ECM contributes to the assembly of individual cells into tissues, affecting this process at both receptor and cytoskeletal levels. Adhesion-mediated signaling, based on the cells’ capacity to sense the chemical and physical properties of the matrix, affects both global cell physiology and local molecular scaffolding of the adhesion sites. The molecular interactions within the adhesion site stimulate, in turn, the signaling process, by clustering together the structural and signaling components of the adhesome.We will also describe the functional molecular architecture of cell–matrix adhesions, highlighting the structure–function relationships between the numerous components of cell adhesions that mediate or modulate numerous cell adhesive, migratory, and regulatory processes. We will discuss the mechanisms underlying the scaffolding and sensing processes generated at integrin-mediated adhesions, considering them along two major multiscale conceptual trajectories: molecular complexity and time—that is, a hierarchy of complexity that spans the range from molecules to multimolecular complexes in mature adhesions, as well as the temporal progression of structures during the assembly and maturation of matrix adhesions, from initial cell–matrix recognition to the formation, maturation, and reorganization of cytoskeleton-associated matrix adhesions.  相似文献   

5.
6.
Classical cadherins mediate specific adhesion at intercellular adherens junctions. Interactions between cadherin ectodomains from apposed cells mediate cell–cell contact, whereas the intracellular region functionally links cadherins to the underlying cytoskeleton. Structural, biophysical, and biochemical studies have provided important insights into the mechanism and specificity of cell–cell adhesion by classical cadherins and their interplay with the cytoskeleton. Adhesive binding arises through exchange of β strands between the first extracellular cadherin domains (EC1) of partner cadherins from adjacent cells. This “strand-swap” binding mode is common to classical and desmosomal cadherins, but sequence alignments suggest that other cadherins will bind differently. The intracellular region of classical cadherins binds to p120 and β-catenin, and β-catenin binds to the F-actin binding protein α-catenin. Rather than stably bridging β-catenin to actin, it appears that α-catenin actively regulates the actin cytoskeleton at cadherin-based cell–cell contacts.Cadherins constitute a large family of cell surface proteins, many of which participate in Ca2+-dependent cell adhesion that plays a fundamental role in the formation of solid tissues (Takeichi 1995; Tepass 1999; Gumbiner 2005). Many events in the development of multicellular assemblies are associated with changes in cadherin expression (Takeichi 1995; Honjo et al. 2000; Price et al. 2002). Expression of particular cadherins often correlates with formation of discrete tissue structures, and in mature tissues discrete cell layers or other cell assemblies are often demarcated by particular cadherins (Gumbiner 1996). Conversely, down-regulation or loss of cadherins correlates with an increased metastatic potential of the affected cells that arises from the loss of their adhesive properties (Hajra and Fearon 2002; Gumbiner 2005).The cadherins of vertebrates, and some of their invertebrate homologs, are the most highly characterized. “Classical” cadherins, associated with the adherens junction, and the closely related desmosomal cadherins feature an amino-terminal extracellular region or ectodomain that is followed by a transmembrane anchor and a carboxy-terminal intracellular region. Interactions between ectodomains on apposed cells mediate specific cell–cell contacts, whereas the intracellular region functionally links cadherins to the underlying cytoskeleton. This article focuses on structural, biophysical, and biochemical studies that have provided important mechanistic insights into the specificity of cell–cell adhesion and its interplay with the cytoskeleton (see also Meng and Takeichi 2009; Cavey and Lecuit 2009).  相似文献   

7.
The extracellular matrix (ECM) and its receptors make diverse contributions to development. The ECM comes in a variety of forms, including the more “standard” ECM that is internal to the animal and on the basal side of epithelial sheets, as well as the apical ECM, which is especially elaborated in the invertebrates to form the exoskeleton. ECM proteins accumulate adjacent to particular target tissues in the developing animal by a variety of mechanisms: local synthesis in the target tissue; local synthesis by migrating cells; and secretion from a distant source and capture by the target tissue. The diverse developmental functions of the ECM are discussed, including the generation of a road for cell migration, creation of morphogenetic checkpoints for differentiation, modulation of morphogen gradients, insulation of organs, gluing together cell layers, and providing structure for the organism.This article will discuss the many functions of the ECM that are important in animal development. First, I will discuss the production of ECM in the developing animal (see Adams and Lawler 2011, Barros et al. 2011; Chiquet-Ehrismann and Tucker 2011; Schwarzbauer and DeSimone 2011; Watt and Fujiwara 2011; Wickstrom et al. 2011; Yurchenco 2011), and then describe briefly the range of ways that the ECM contributes to development. Given the space constraints of this article, I will not attempt to be exhaustive in my coverage, but instead will seek to identify examples that show the different kinds of ECM function that have been revealed from studies of development. My examples will be focused on developmental processes shared between invertebrates and vertebrates, as many vertebrate-specific roles of the ECM are covered in other references that are part of this subject collection. The somewhat different compositions, arrangements, and developmental changes in invertebrate ECMs provide an illuminating perspective from which to consider ECM function more generally.  相似文献   

8.
9.
The Wnt pathway is a major embryonic signaling pathway that controls cell proliferation, cell fate, and body-axis determination in vertebrate embryos. Soon after egg fertilization, Wnt pathway components play a role in microtubule-dependent dorsoventral axis specification. Later in embryogenesis, another conserved function of the pathway is to specify the anteroposterior axis. The dual role of Wnt signaling in Xenopus and zebrafish embryos is regulated at different developmental stages by distinct sets of Wnt target genes. This review highlights recent progress in the discrimination of different signaling branches and the identification of specific pathway targets during vertebrate axial development.Wnt pathways play major roles in cell-fate specification, proliferation and differentiation, cell polarity, and morphogenesis (Clevers 2006; van Amerongen and Nusse 2009). Signaling is initiated in the responding cell by the interaction of Wnt ligands with different receptors and coreceptors, including Frizzled, LRP5/6, ROR1/2, RYK, PTK7, and proteoglycans (Angers and Moon 2009; Kikuchi et al. 2009; MacDonald et al. 2009). Receptor activation is accompanied by the phosphorylation of Dishev-elled (Yanagawa et al. 1995), which appears to transduce the signal to both the cell membrane and the nucleus (Cliffe et al. 2003; Itoh et al. 2005; Bilic et al. 2007). Another common pathway component is β-catenin, an abundant component of adherens junctions (Nelson and Nusse 2004; Grigoryan et al. 2008). In response to signaling, β-catenin associates with T-cell factors (TCFs) and translocates to the nucleus to stimulate Wnt target gene expression (Behrens et al. 1996; Huber et al. 1996; Molenaar et al. 1996).This β-catenin-dependent activation of specific genes is often referred to as the “canonical” pathway. In the absence of Wnt signaling, β-catenin is destroyed by the protein complex that includes Axin, GSK3, and the tumor suppressor APC (Clevers 2006; MacDonald et al. 2009). Wnt proteins, such as Wnt1, Wnt3, and Wnt8, stimulate Frizzled and LRP5/6 receptors to inactivate this β-catenin destruction complex, and, at the same time, trigger the phosphorylation of TCF proteins by homeodomain-interacting protein kinase 2 (HIPK2) (Hikasa et al. 2010; Hikasa and Sokol 2011). Both β-catenin stabilization and the regulation of TCF protein function by phosphorylation appear to represent general strategies that are conserved in multiple systems (Sokol 2011). Thus, the signaling pathway consists of two branches that together regulate target gene expression (Fig. 1).Open in a separate windowFigure 1.Conserved Wnt pathway branches and components. In the absence of Wnt signals, glycogen synthase kinase 3 (GSK3) binds Axin and APC to form the β-catenin destruction complex. Some Wnt proteins, such as Wnt8 and Wnt3a, stimulate Frizzled and LRP5/6 receptors to inhibit GSK3 activity and stabilize β-catenin (β-cat). Stabilized β-cat forms a complex with T-cell factors (e.g., TCF1/LEF1) to activate target genes. Moreover, GSK3 inhibition leads to target gene derepression by promoting TCF3 phosphorylation by homeodomain-interacting protein kinase 2 (HIPK2) through an unknown mechanism, for which β-catenin is required as a scaffold. This phosphorylation results in TCF3 removal from target promoters and gene activation. Other Wnt proteins, such as Wnt5a and Wnt11, use distinct receptors such as ROR2 and RYK, in addition to Frizzled, to control the the cytoskeletal organization through core planar cell polarity (PCP) proteins, small GTPases (Rho/Rac/Cdc42), and c-Jun amino-terminal kinase (JNK).Other Wnt proteins, such as Wnt5a or Wnt11, strongly affect the cytoskeletal organization and morphogenesis without stabilizing β-catenin (Torres et al. 1996; Angers and Moon 2009; Wu and Mlodzik 2009). These “noncanonical” ligands do not influence TCF3 phosphorylation (Hikasa and Sokol 2011), but may use distinct receptors such as ROR1/2 and RYK instead of or in addition to Frizzled (Hikasa et al. 2002; Lu et al. 2004; Mikels and Nusse 2006; Nishita et al. 2006, 2010; Schambony and Wedlich 2007; Grumolato et al. 2010; Lin et al. 2010; Gao et al. 2011). In such cases, signaling mechanisms are likely to include planar cell polarity (PCP) components, such as Vangl2, Flamingo, Prickle, Diversin, Rho GTPases, and c-Jun amino-terminal kinases (JNKs), which do not directly affect β-catenin stability (Fig. 1) (Sokol 2000; Schwarz-Romond et al. 2002; Schambony and Wedlich 2007; Komiya and Habas 2008; Axelrod 2009; Itoh et al. 2009; Tada and Kai 2009; Sato et al. 2010; Gao et al. 2011). This simplistic dichotomy of the Wnt pathway does not preclude some Wnt ligands from using both β-catenin-dependent and -independent routes in a context-specific manner.Despite the existence of many pathway branches, only the β-catenin-dependent branch has been implicated in body-axis specification. Recent experiments in lower vertebrates have identified additional pathway components and targets and provided new insights into the underlying mechanisms.  相似文献   

10.
Integrins are large, membrane-spanning, heterodimeric proteins that are essential for a metazoan existence. All members of the integrin family adopt a shape that resembles a large “head” on two “legs,” with the head containing the sites for ligand binding and subunit association. Most of the receptor dimer is extracellular, but both subunits traverse the plasma membrane and terminate in short cytoplasmic domains. These domains initiate the assembly of large signaling complexes and thereby bridge the extracellular matrix to the intracellular cytoskeleton. To allow cells to sample and respond to a dynamic pericellular environment, integrins have evolved a highly responsive receptor activation mechanism that is regulated primarily by changes in tertiary and quaternary structure. This review summarizes recent progress in the structural and molecular functional studies of this important class of adhesion receptor.The name “integrin” was suggested for an integral membrane protein complex first characterized in 1986 (Tamkun et al. 1986). The name was devised because the protein identified linked the extracellular matrix to the cytoskeleton (early developments in this field have been well described [Hynes 2004]). In the 25 years since that first characterization, a vast amount of work has been performed, with consequent increased understanding. The essential role of integrins in tissue organization and cell development, their signal transduction mechanisms (from outside to in and inside to out!), and their potential as therapeutic targets is now established. In this article, we provide an overview of the structure of integrins, the conformational changes that determine activation state, and the mechanisms of ligand binding.  相似文献   

11.
One of the early surprises in the study of cell adhesion was the discovery that β-catenin plays dual roles, serving as an essential component of cadherin-based cell–cell adherens junctions and also serving as the key regulated effector of the Wnt signaling pathway. Here, we review our current model of Wnt signaling and discuss how recent work using model organisms has advanced our understanding of the roles Wnt signaling plays in both normal development and in disease. These data help flesh out the mechanisms of signaling from the membrane to the nucleus, revealing new protein players and providing novel information about known components of the pathway.Modern biomedical science is a partnership between scientists studying basic cell and developmental processes in model systems and clinicians exploring the basis of human disease. Few fields exemplify this better than Wnt signaling, born 22 years ago with the realization that the oncogene int1 and the Drosophila developmental patterning gene wingless (wg) are homologs (Cabrera et al. 1987; Rijsewijk et al. 1987). Additional connections further fueled research. Drosophila Armadillo (Arm), a component of the Wg pathway, is the homolog of the cell junction proteins β-catenin (βcat) and plakoglobin (McCrea et al. 1991; Peifer et al. 1992; Peifer and Wieschaus 1990) joining Wnt signaling and cadherin-based cell adhesion, a connection we still do not fully understand (see Heuberger and Birchmeier 2009). Adenomatous polyposis coli (APC), the tumor suppressor mutated in most colon cancers, binds βcat and is a key regulator of Wnt signaling (Rubinfeld et al. 1993; Su et al. 1993), putting the Wnt field even more squarely in the center of cancer research. Here, we outline recent advances in understanding Wnt signaling, casting new light on these critical regulators of development, homeostasis, and disease.  相似文献   

12.
13.
Although the actin cytoskeleton and T-cell receptor (TCR) signaling complexes are seemingly distinct molecular structures, they are tightly integrated in T cells. The signaling pathways initiated by TCRs binding to peptide MHC complexes are extensively influenced by the actin cytoskeletal activities of the motile phase before TCR signaling, the signalosome scaffolding function of the cytoskeleton, and the translocation of signaling clusters that precedes the termination of signaling at these complexes. As these three successive phases constitute essentially all the steps consequent to immune synapse formation, it has become clear that the substantial physical forces and signaling interactions generated by the actin cytoskeleton dominate the signaling life cycle of TCR signalosomes. We discuss the contributions of the actin cytoskeleton to TCR signaling phases and model some remaining questions about how specific cytoskeletal factors regulate TCR signaling outcomes.The activation of T cells is controlled primarily by T-cell receptors (TCRs) interacting with peptide-loaded major histocompatibility complexes (pMHCs) as T cells scan the surface of antigen presenting cells (APCs). Because T cells are continuously motile cells that transit through lymph nodes in their surveillance, it is clear that TCR triggering must occur within the context of physical forces that might rapidly separate TCRs from agonist pMHCs. Moreover, crawling T cells do not truly come to rest at the surfaces of APCs following TCR engagement. Instead, they continuously extend protrusions over APCs and move along the surface of their partner (Gunzer et al. 2000). In their initial encounters with antigen-bearing dendritic cells (DCs), T cells also often rapidly couple and uncouple on the order of minutes, rather than dwelling for extended periods of time on single DCs (Gunzer et al. 2000; Mempel et al. 2004). This dynamic coupling allows T cells to quickly sample a large proportion of the total APC membrane pool in search of their cognate antigen. Still, these transient contacts are productive—they induce calcium fluxes and the expression of markers of activated T cells—indicating that TCR signalosome outputs can be initiated in mere minutes and survive the dissolution of contacts, even under the mechanical stress of cytoskeletal remodeling.TCR signaling requires the dynamic recruitment of a macromolecular complex of kinases, scaffolding molecules, and other signaling effectors to a triggered TCR. Assembly of this macromolecular signaling complex must be very sensitive and occur rapidly, or there is a risk that the TCR will release the pMHC ligand, and the T cell will fail to register the antigen hit. Conversely, the signalosome assembly mechanism needs to discriminate against TCRs interacting transiently with a vast array of pMHCs presenting nonagonist peptides. Viewed in this manner, a scheme that rapidly dissociates TCRs from MHCs loaded with endogenous peptide, freeing them to rebind and test other MHCs, is desirable. It is notable that several TCR signaling factors carry binding sites for actin binding proteins or actin itself (Rozdzial et al. 1995; Zhang et al. 1999; Zeng et al. 2003; Phee et al. 2005; Gomez et al. 2006). Through these actin-associated factors, agonist-triggered TCRs rapidly assemble stabilized signaling platforms that survive mechanical disruption.In concert with adhesive integrin interactions and costimulatory receptor signaling, TCRs orchestrate a reorganization of the T-cell plasma membrane that may begin with a handful of receptors and eventually encompasses the entire contact face with the APC (some 50–100 µm2). TCRs first aggregate into micron scale clusters of TCRs, then flow to the center of the contact face, generating the central supramolecular activating complex (cSMAC) of the immune synapse (Monks et al. 1998; Grakoui et al. 1999; Krummel et al. 2000). Underscoring the importance of the cytoskeleton, the actin depolymerizing toxins latrunculin A and cytochalasin D are potent inhibitors of T-cell activation and block both TCR microcluster formation and cSMAC coalescence (Wulfing et al. 1998; Grakoui et al. 1999; Krummel et al. 2000; Varma et al. 2006). Ultimately, it is the coordination of the local interactions between receptors and effectors with the cell morphological level rearrangements that determines the nature and magnitude of T-cell responses to pathogens. Regulation of TCR signaling lifecycles and T-cell responses, therefore, falls squarely on the actin cytoskeleton.  相似文献   

14.
The response of innate immune cells to growth factors, immune complexes, extracellular matrix proteins, cytokines, pathogens, cellular damage, and many other stimuli is regulated by a complex net of intracellular signal transduction pathways. The majority of these pathways are either initiated or modulated by Src-family or Syk tyrosine kinases present in innate cells. The Src-family kinases modulate the broadest range of signaling responses, including regulating immunoreceptors, C-type lectins, integrins, G-protein-coupled receptors, and many others. Src-family kinases also modulate the activity of other kinases, including the Tec-family members as well as FAK and Pyk2. Syk kinase is required for initiation of signaling involving receptors that utilize immunoreceptor tyrosine activation (ITAM) domains. This article reviews the major activating and inhibitory signaling pathways regulated by these cytoplasmic tyrosine kinases, illuminating the many examples of signaling cross talk between pathways.Innate immune cells, including macrophages, dendritic cells, granulocytes, and mast cells, function as the first line of defense against pathogens. These cells use a dizzying array of cell-surface receptors, which are connected to an equally complicated intracellular signal transduction network, to sense pathogen molecules and then orchestrate the appropriate immune response. Among the intracellular signaling molecules that are most crucial for innate immune cells are the cytoplasmic tyrosine kinases. Two major kinase families that operate in the proximal intracellular signaling pathways in innate cells are the Src-family kinases and the Syk-ZAP70 family. A third family of kinases, the Tek family, also have important roles in innate cells. They are not discussed in detail in this article, but are reviewed elsewhere in articles on the subject.There are eight members of the Src family; innate immune cells primarily express Hck, Fgr, Lyn, and to a lesser extent, Src (Lowell 2004). The Syk-ZAP70 family has only two members and only Syk is found in innate cells. Most innate cell types express the same spectrum of kinases with some specific cellular differences. For example, mast cells express a broader range of Src-family kinases than macrophages or dendritic cells (Colgan and Hankel 2010). In general, Src-family and Syk kinases tend to operate together in signaling pathways, with the Src-family being “upstream” or activated first in response to pathogen detection. These enzymes then communicate downstream to Tec-family members. The Tec-family kinases expressed in innate cells include Btk, Bmx, and Tec (Koprulu and Ellmeier 2009; Tohyama and Yamamura 2009). Additionally, Src-family kinases activate yet another family of PTKs, the FAK/Pyk2 tyrosine kinases, which play a major role in integrin signaling (Hauck et al. 2000).Though primarily studied in activating pathways, Src-family and Syk kinases also activate inhibitory signaling pathways (Nimmerjahn and Ravetch 2008). In many situations, inhibitory signaling often overrides the activating signal. Pathways can also be initiated at different times or rates. Finally, to add even more complexity, activating and inhibitory pathways often interact indirectly, for example, through the production of cytokines and growth factors and not through direct intracellular biochemical interactions; Hence the term signaling “cross talk,” which now appears commonly in the literature (O''Neill 2008; Ivashkiv 2009; Page et al. 2009).  相似文献   

15.
The problem of recognizing and disposing of non-self-organisms, whether for nutrients or defense, predates the evolution of multicellularity. Accordingly, the function of the innate immune system is often intimately associated with fundamental aspects of cell biology. Here, we review our current understanding of the links between cell biology and pattern-recognition receptors of the innate immune system. We highlight the importance of receptor localization for the detection of microbes and for the initiation of antimicrobial signaling pathways. We discuss examples that illustrate how pattern-recognition receptors influence, and are influenced by, the general membrane trafficking machinery of mammalian cells. In the future, cell biological analysis likely will rival pure genetic analysis as a tool to uncover fundamental principles that govern host–microbe interactions.The innate immune system uses families of pattern-recognition receptors (PRRs) to recognize diverse microbial ligands (Janeway 1989; Janeway and Medzhitov 2002). During infection, these receptors provide signals that up-regulate general antimicrobial features of the innate immune system as well as instruct and initiate adaptive immunity (Iwasaki and Medzhitov 2010). A significant challenge faced by innate immune recognition is the reliable detection of highly diverse, rapidly evolving microbial organisms, many of which possess virulence mechanisms that enable survival within distinct host niches. Moreover, recognition must be linked to induction of contextual signals appropriate for the type of infection. The specificity, signal transduction, and cell biology of PRRs have evolved under these selective pressures to enable broad recognition of microbes within each host niche.Although the collection of PRRs is decidedly less diverse than antigen receptors of the adaptive immune system, the list of players has grown considerably over the past decade (Kawai and Akira 2010). If one classifies these receptors based on common structure and functional domains, then six families emerge: Toll-like receptors (TLRs), C-type lectin receptors (CLRs), RIG-I-like receptors (RLRs), AIM-like receptors (ALRs), Nod-like receptors (NLRs), and OAS-like receptors (OLRs) (Geijtenbeek and Gringhuis 2009; Kawai and Akira 2010; Rathinam and Fitzgerald 2011; Lamkanfi and Dixit 2012; Kranzusch et al. 2013). Collectively, these receptors bind a diverse array of targets, including lipoproteins, polysaccharides, nucleic acids, carbohydrate structures, and a few highly conserved microbial proteins. These ligands are typically shared across large microbial classes, which facilitate broad recognition with such a limited number of PRRs. Moreover, alteration or masking of these ligands to avoid PRR activation often results in reduced microbial fitness.The molecular recognition challenge faced by PRRs is all the more complex when one considers the need to detect microbes within distinct subcellular niches. Microbes can be extracellular or intracellular within membrane-bound organelles, within the cytosol, or in the nucleus. In addition, both the innate and adaptive immune mechanisms appropriate for eliminating microbes within these distinct environments are quite distinct, so it is vital that PRR signaling communicate the location of a microbe as well its nature. We now understand that members of the PRR families highlighted above localize to distinct subcellular compartments, and, in some cases, localization can change in a dynamic fashion that regulates or influences recognition and signaling. Moreover, in some cases, signal transduction and resulting gene induction can be dramatically influenced by the organelle from which signaling initiates. Thus, the innate immune system has harnessed the organization inherent to cells as a means of achieving regulation and signaling specificity. Activation of PRRs can also feed back on basic cell biological processes, such as phagocytosis and autophagy, to enhance or accelerate the response to microbial infection.In the following sections, we discuss these links between cell biology and PRRs of mammalian innate immunity. Our discussions of PRR function and signal transduction will be limited to this theme, as a result, in part, of space constraints but also because in-depth reviews of each PRR family have appeared elsewhere. For discussion purposes, we have grouped the transmembrane PRRs together and the cytosolic PRRs together.  相似文献   

16.
Epithelia form physical barriers that separate the internal milieu of the body from its external environment. The biogenesis of functional epithelia requires the precise coordination of many cellular processes. One of the key events in epithelial biogenesis is the establishment of cadherin-dependent cell–cell contacts, which initiate morphological changes and the formation of other adhesive structures. Cadherin-mediated adhesions generate intracellular signals that control cytoskeletal reorganization, polarity, and vesicle trafficking. Among such signaling pathways, those involving small GTPases play critical roles in epithelial biogenesis. Assembly of E-cadherin activates several small GTPases and, in turn, the activated small GTPases control the effects of E-cadherin-mediated adhesions on epithelial biogenesis. Here, we focus on small GTPase signaling at E-cadherin-mediated epithelial junctions.Cell–cell adhesions are involved in a diverse range of physiological processes, including morphological changes during tissue development, cell scattering, wound healing, and synaptogenesis (Adams and Nelson 1998; Gumbiner 2000; Halbleib and Nelson 2006; Takeichi 1995; Tepass et al. 2000). In epithelial cells, cell–cell adhesions are classified into three kinds of adhesions: adherens junction, tight junction, and desmosome (for more details, see Meng and Takeichi 2009, Furuse 2009, and Delva et al. 2009, respectively). A key event in epithelial polarization and biogenesis is the establishment of cadherin-dependent cell–cell contacts. Cadherins belong to a large family of adhesion molecules that require Ca2+ for their homophilic interactions (Adams and Nelson 1998; Blanpain and Fuchs 2009; Gumbiner 2000; Hartsock and Nelson 2008; Takeichi 1995; Tepass et al. 2000). Cadherins form transinteraction on the surface of neighboring cells (for details, see Shapiro and Weis 2009). For the development of strong and rigid adhesions, cadherins are clustered concomitantly with changes in the organization of the actin cytoskeleton (Tsukita et al. 1992). Classical cadherins are required, but not sufficient, to initiate cell–cell contacts, and other adhesion protein complexes subsequently assemble (for details, see Green et al. 2009). These complexes include the tight junction, which controls paracellular permeability, and desmosomes, which support the structural continuum of epithelial cells. A fundamental problem is to understand how these diverse cellular processes are regulated and coordinated. Intracellular signals, generated when cells attach with one another, mediate these complicated processes.Several signaling pathways upstream or downstream of cadherin-mediated cell–cell adhesions have been identified (Perez-Moreno et al. 2003) (see also McCrea et al. 2009). Among these pathways, small GTPases including the Rho and Ras family GTPases play critical roles in epithelial biogenesis and have been studied extensively. Many key morphological and functional changes are induced when these small GTPases act at epithelial junctions, where they mediate an interplay between cell–cell adhesion molecules and fundamental cellular processes including cytoskeletal activity, polarity, and vesicle trafficking. In addition to these small GTPases, Ca2+ signaling and phosphorylation of cadherin complexes also play pivotal roles in the formation and maintenance of cadherin-mediated adhesions. Here, we focus on signaling pathways involving the small GTPases in E-cadherin-mediated cell–cell adhesions. Other signaling pathways are described in recent reviews (Braga 2002; Fukata and Kaibuchi 2001; Goldstein and Macara 2007; McLachlan et al. 2007; Tsukita et al. 2008; Yap and Kovacs 2003; see also McCrea et al. 2009).  相似文献   

17.
18.
19.
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   

20.
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   

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