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 62 毫秒
1.
Short interfering RNAs (siRNAs) are a class of regulatory effectors that enforce gene silencing through formation of RNA duplexes. Although progress has been made in identifying the capabilities of siRNAs in silencing foreign RNA and transposable elements, siRNA functions in endogenous gene regulation have remained mysterious. In certain organisms, siRNA biosynthesis involves novel enzymes that act as RNA-directed RNA polymerases (RdRPs). Here we analyze the function of a Caenorhabditis elegans RdRP, RRF-3, during spermatogenesis. We found that loss of RRF-3 function resulted in pleiotropic defects in sperm development and that sperm defects led to embryonic lethality. Notably, sperm nuclei in mutants of either rrf-3 or another component of the siRNA pathway, eri-1, were frequently surrounded by ectopic microtubule structures, with spindle abnormalities in a subset of the resulting embryos. Through high-throughput small RNA sequencing, we identified a population of cellular mRNAs from spermatogenic cells that appear to serve as templates for antisense siRNA synthesis. This set of genes includes the majority of genes known to have enriched expression during spermatogenesis, as well as many genes not previously known to be expressed during spermatogenesis. In a subset of these genes, we found that RRF-3 was required for effective siRNA accumulation. These and other data suggest a working model in which a major role of the RRF-3/ERI pathway is to generate siRNAs that set patterns of gene expression through feedback repression of a set of critical targets during spermatogenesis.REPRESSION of gene expression by small RNAs of ∼20–30 nt in length is important for many aspects of multicellular eukaryotic development. A variety of classes of small RNA with distinct structural features, modes of biogenesis, and biological functions have been identified (reviewed in Hutvagner and Simard 2008). We are particularly interested in a class of small RNAs, called endogenous short interfering RNAs (siRNAs), that are similar to intermediates in exogenously triggered RNA interference (RNAi) in their perfect complementarity to mRNA targets. High-throughput sequencing technology has provided a valuable tool for characterization of endogenous siRNA populations from many diverse sources, including mouse embryonic stem cells (Babiarz et al. 2008), Drosophila heads (Ghildiyal et al. 2008), and Arabidopsis pollen (Slotkin et al. 2009). These siRNAs have been proposed to function in the regulation of both cellular processes and genome defense through downregulation of gene expression. Caenorhabditis elegans, like plants and fungi, utilizes RNA-copying enzymes called RNA-directed RNA polymerases (RdRPs) as part of the RNAi machinery (Smardon et al. 2000; Sijen et al. 2001). While two of the C. elegans RdRPs are nonessential (RRF-1 and RRF-2), mutations in either of the remaining two (EGO-1 or RRF-3) lead to fertility defects (Smardon et al. 2000; Simmer et al. 2002). RRF-3 is functionally distinct from EGO-1 in that the RRF-3 requirement in fertility is temperature dependent. In addition, RRF-3 activity has an inhibitory effect on exogenously triggered RNAi (resulting in an ERI, or enhanced RNAi, mutant phenotype in rrf-3 mutants). Mutants lacking either RRF-3 or another ERI factor, ERI-1, have been used as experimental tools because of their enhanced sensitivity in RNAi-based screens. One proposed mechanism for the enhancement in RNAi in rrf-3 and eri mutants has been a competition for cofactors between the exogenously triggered RNAi pathway and an endogenous RNAi pathway. Consistent with this hypothesis, siRNAs corresponding to several genes have been shown by Northern analysis to depend upon RRF-3 and other ERI factors for their accumulation (Duchaine et al. 2006; Lee et al. 2006; Yigit et al. 2006). Global microarray analyses have also been undertaken to identify messenger RNAs whose expression is affected by RRF-3 and ERI-1 (Lee et al. 2006; Asikainen et al. 2007).A functional significance of the RRF-3/ERI pathway has been inferred by the inability of rrf-3, eri-1, eri-3, and eri-5 mutant strains to propagate at a high growth temperature (Simmer et al. 2002; Duchaine et al. 2006). Rather than producing temperature-sensitive mutant protein effects, RRF-3 and other ERI proteins are thought to act in a temperature-sensitive process, as evidenced by the predicted truncated and presumed nonfunctional protein fragments that would result from the available deletion alleles and by their shared temperature-sensitive phenotypes. rrf-3 mutant animals have been observed to exhibit X-chromosome missegregation (Simmer et al. 2002) and an unusual persistence of a chromatin mark on the X chromosome during male spermatogenesis (Maine et al. 2005). X-chromosome missegregation and defective spermatogenesis have been referred to in previous studies of eri-1 (Kennedy et al. 2004) and eri-3 and eri-5 (Duchaine et al. 2006). Furthermore, eri-3 mutant sterility can be rescued by insemination with wild-type sperm (Duchaine et al. 2006).Here we investigated the role of RRF-3 during spermatogenesis. We found defects evident at multiple stages, including after fertilization, where defects in rrf-3 mutant sperm can produce subsequent nonviable embryos. By using high-throughput sequencing, we characterized a large population of siRNAs present in spermatogenic cells and found a strong enrichment for antisense siRNAs from genes with known mRNA expression during spermatogenesis. While the majority of siRNA production during spermatogenesis does not require RRF-3, we found a set of genes for which siRNA production was dependent upon RRF-3. Existing data indicate increased expression for these genes in rrf-3 and/or eri-1 mutants. Taken together, our analyses suggest a working model in which the RRF-3/ERI pathway generates siRNAs that downregulate specific genes during spermatogenesis, with this regulation playing a key role in generating functional sperm.  相似文献   

2.
The nerve-cell cytoskeleton is essential for the regulation of intrinsic neuronal activity. For example, neuronal migration defects are associated with microtubule regulators, such as LIS1 and dynein, as well as with actin regulators, including Rac GTPases and integrins, and have been thought to underlie epileptic seizures in patients with cortical malformations. However, it is plausible that post-developmental functions of specific cytoskeletal regulators contribute to the more transient nature of aberrant neuronal activity and could be masked by developmental anomalies. Accordingly, our previous results have illuminated functional roles, distinct from developmental contributions, for Caenorhabditis elegans orthologs of LIS1 and dynein in GABAergic synaptic vesicle transport. Here, we report that C. elegans with function-altering mutations in canonical Rac GTPase-signaling-pathway members demonstrated a robust behavioral response to a GABAA receptor antagonist, pentylenetetrazole. Rac mutants also exhibited hypersensitivity to an acetylcholinesterase inhibitor, aldicarb, uncovering deficiencies in inhibitory neurotransmission. RNA interference targeting Rac hypomorphs revealed synergistic interactions between the dynein motor complex and some, but not all, members of Rac-signaling pathways. These genetic interactions are consistent with putative Rac-dependent regulation of actin and microtubule networks and suggest that some cytoskeletal regulators cooperate to uniquely govern neuronal synchrony through dynein-mediated GABAergic vesicle transport in C. elegans.EPILEPSY affects 1–2% of the world population and is associated with imbalances between excitatory and inhibitory neurotransmission in the brain (Locke et al. 2009). In particular, interneurons expressing gamma-aminobutyric acid (GABA), the principal inhibitory neurotransmitter in the human brain, are essential for normal neuronal synchronization and maintenance of a seizure threshold in humans (Cossette et al. 2002), rodents (Delorey et al. 1998), and zebrafish (Baraban et al. 2005). A failure of the brain to properly regulate neuronal synchrony can result from ion channel defects (Xu and Clancy 2008), neuropeptide depletion (Brill et al. 2006), brain malformations (Patel et al. 2004), interneuron loss (Cobos et al. 2005), and/or synaptic vesicle recycling failure (Di Paolo et al. 2002), all of which may be caused by disrupting the nerve-cell cytoskeleton. Therefore, further exploration of putative links between cytoskeletal components and neurotransmission may accelerate development of novel therapeutics for epilepsy.Epilepsy associated with cytoskeletal dysfunction often has a developmental basis (Di Cunto et al. 2000; Wenzel et al. 2001; Keays et al. 2007). For example, mutations in LIS1, a dynein motor complex regulator, lead to classical lissencephaly, which is characterized by neuronal migration defects, a lack of convolutions in the brain, mental retardation, and epileptic seizures (Lo Nigro et al. 1997). Yet, observations that lissencephaly-associated seizures worsen after neuronal migration ceases, while LIS1 expression persists, imply that LIS1 also acts in the adult brain (Cardoso et al. 2002).We previously reported that C. elegans with a predicted null mutation (t1550) in lis-1, the worm ortholog of human LIS1, exhibited synaptic vesicle misaccumulations, but not neuronal migration or axon-pathfinding defects, in GABAergic motor neurons. We also observed anterior “epileptic-like” convulsions, which were intense, frequent, and repetitive, with lis-1(t1550) homozygotes in the presence of pentylenetetrazole (PTZ; Williams et al. 2004), an epileptogenic GABAA receptor antagonist (Huang et al. 2001; Fernandez et al. 2007). PTZ sensitivity was also increased in heterozygous lis-1(t1550) mutants following RNA interference (RNAi) against worm orthologs of associated cortical malformation genes, such as cdk-5 and nud-2, which are known to interact with LIS1 and the dynein motor complex. Depletion of these gene products was coincident with dynein-mediated synaptic vesicle transport defects, not with architectural defects, in GABAergic motor neurons (Locke et al. 2006).Plausible functional interactions among LIS-1, dynein, and Rac GTPases (Rehberg et al. 2005; Kholmanskikh et al. 2006) have not been explored in an intact adult nervous system. C. elegans is ideal for characterizing these interactions due to the availability of weak and strong Rac pathway mutants (Lundquist et al. 2001; Poinat et al. 2002; Lucanic et al. 2006), a comprehensive RNAi library (Kamath et al. 2003), and GFP-based neuronal markers. Here, we combine these tools with pharmacological modifiers of neuronal activity and establish an experimental paradigm that reveals a novel regulatory pathway. This pathway is composed of integrins at the plasma membrane that signal through Racs to dynein-associated proteins, which function to coordinate synaptic vesicle transport in larval and adult GABAergic motor neurons.  相似文献   

3.
4.
On the basis of the free radical and rate of living theories of aging, it has been proposed that decreased metabolism leads to increased longevity through a decreased production of reactive oxygen species (ROS). In this article, we examine the relationship between mitochondrial energy metabolism and life span by using the Clk mutants in Caenorhabditis elegans. Clk mutants are characterized by slow physiologic rates, delayed development, and increased life span. This phenotype suggests that increased life span may be achieved by decreasing energy expenditure. To test this hypothesis, we identified six novel Clk mutants in a screen for worms that have slow defecation and slow development and that can be maternally rescued. Interestingly, all 11 Clk mutants have increased life span despite the fact that slow physiologic rates were used as the only screening criterion. Although mitochondrial function is decreased in the Clk mutants, ATP levels are normal or increased, suggesting decreased energy utilization. To determine whether the longevity of the Clk mutants results from decreased production of ROS, we examined sensitivity to oxidative stress and oxidative damage. We found no evidence for systematically increased resistance to oxidative stress or decreased oxidative damage in the Clk mutants despite normal or elevated levels of superoxide dismutases. Overall, our findings suggest that decreased energy metabolism can lead to increased life span without decreased production of ROS.MUTATIONS in clk-1 have been shown to increase longevity in both worms and mice, suggesting that these mutations affect an evolutionarily conserved mechanism of life span extension (Lakowski and Hekimi 1996; Liu et al. 2005; Lapointe et al. 2009). The CLK-1 protein encodes a hydroxylase involved in the synthesis of ubiquinone (Ewbank et al. 1997), a multifunctional, lipid-like molecule that transfers electrons in the electron transport chain and may also act as an intracellular antioxidant (Maroz et al. 2009). clk-1 was originally identified in worms in a screen for maternally rescued mutations that result in abnormal development and behavior. In addition to slow development and slow defecation, clk-1 mutants show decreased brood size, a decreased rate of thrashing, and a decreased rate of pharyngeal pumping (Wong et al. 1995). It was a surprise, however, that clk-1 worms also displayed extended longevity, because, at the time that it was discovered, only two other mutants, age-1 and daf-2, with very different phenotypes, had been found to extend longevity (Friedman and Johnson 1988; Kenyon et al. 1993).It is currently uncertain how mutations in clk-1 result in the overall slowing of development and physiologic rates as well as an extended life span. One classic theory of aging, called the rate of living theory, postulates the existence of a link between energy metabolism and aging (Pearl 1922; Speakman 2005). This theory proposes that what determines the life span of an organism is the rate at which it produces and uses energy at the cellular level. Thus, the fact that clk-1 worms exhibit slow physiologic rates and development suggests a decrease in the rate that these worms utilize energy, and, by the rate of living theory, this could account for their long life span.In support of the rate of living theory, the loss of clk-1 has been shown to result in decreased whole-worm oxygen consumption (Felkai et al. 1999; Yang et al. 2007) and decreased electron transfer from complex I to complex III in the electron transport chain (Kayser et al. 2004b), although this has not been observed by all investigators (Miyadera et al. 2001). While some reports have suggested that energy consumption is not reduced in clk-1 worms, at least under liquid culture conditions (Braeckman et al. 2002), the observation that clk-1 worms have higher levels of ATP than wild-type worms (Braeckman et al. 1999) suggests a decreased use of energy in clk-1 worms regardless of whether energy production is normal or decreased. It has also been found that clk-1 double-mutant combinations that exhibit slower development than clk-1 worms live even longer than clk-1 worms (Lakowski and Hekimi 1996). In addition, overexpression of clk-1 prevents the slowing of the defecation rate with age, increases mitochondrial function, and decreases life span (Felkai et al. 1999).Drawing on ideas from the free radical theory of aging (Harman 1956), it has been suggested that a possible mechanism underlying the rate of living theory is that decreased metabolism results in a lower rate of production of reactive oxygen species (ROS). As the free radical theory of aging proposes that aging results from the accumulation of molecular damage caused by ROS, then lower ROS production should result in slower aging. In clk-1 worms, it has not been possible to directly measure levels of ROS in vivo; however, measurement of hydrogen peroxide production from submitochondrial particles has demonstrated increased ROS generation in clk-1 mitochondria compared to wild type (Yang et al. 2009). In addition, the superoxide production potential is increased in clk-1 worms compared to wild-type N2 worms (Braeckman et al. 2002). Despite showing increased levels of ROS production, clk-1 worms have been found to have normal or decreased levels of oxidative damage (Kayser et al. 2004a; Yang et al. 2007, 2009) and decreased accumulation of lipofuscin (Braeckman et al. 2002). The decrease in oxidative damage that occurs in spite of increased ROS production likely results from increased antioxidant defenses. In support of this conclusion, sod-2 and sod-3 mRNA are increased in clk-1 worms compared to wild type (Yang et al. 2007).Clearly, the levels of ROS production and antioxidant defense are altered in clk-1 worms and likely contribute to the physiology and life span of these worms. Evidence supporting a role for altered ROS levels in determining the clk-1 phenotype comes from the demonstration that increasing the levels of ROS through decreasing superoxide dismutase expression has been shown to modulate a variety of phenotypes in clk-1 worms (Shibata et al. 2003; Yang et al. 2007). It is important to note, however, that the decrease in oxidative damage in clk-1 worms appears not to contribute to their long life as it is possible to experimentally increase oxidative damage in clk-1 worms beyond wild-type levels without reducing life span (Yang et al. 2007).In addition to clk-1, four other genes have been identified that yield a clk-1-like phenotype (Clk phenotype), which includes slow development, slow defecation, slow pharyngeal pumping, decreased brood size and long life span coupled with maternal rescue (homozygous mutants from heterozygous mothers are phenotypically normal) (Hekimi et al. 1995; Lemieux et al. 2001). The Clk phenotype has been studied in most detail in clk-1 worms (Wong et al. 1995) and, subsequently, with gro-1 (Lemieux et al. 2001), clk-2 (Benard et al. 2001), and tpk-1 worms (de Jong et al. 2004), while clk-3 worms have not been extensively studied [although clk-3 worm energy metabolism and oxygen consumption have been examined (Braeckman et al. 2002; Shoyama et al. 2009)]. Despite the phenotypic similarity of these mutants, the mutations that have been identified thus far have been shown to occur in genes encoding proteins with a wide range of functions with no obvious relationship to one another. gro-1 encodes a tRNA-modifying enzyme (Lemieux et al. 2001), clk-2 encodes a homolog of yeast Tel2p and a regulator of several PI3K-related protein kinases (Ahmed et al. 2001; Benard et al. 2001; Jiang et al. 2003; Takai et al. 2007), and tpk-1 encodes thiamine pyrophosphokinase, which is necessary for the assimilation of thiamine (vitamin B1) (de Jong et al. 2004).All of the Clk mutants that have been identified exhibit slow physiologic rates and increased life span, suggesting that one may be sufficient for the other. To test this hypothesis, we identified six novel Clk mutants and demonstrate that these strains bear all of the characteristic features of the Clk phenotype, including extended longevity. We further show that mitochondrial function is decreased in the Clk mutants but that this decrease does not result in increased resistance to oxidative stress or decreased oxidative damage. Our results provide a plausible explanation for the extended life span observed in the Clk mutants and support aspects of the rate of living theory of aging while casting further doubt on the free radical theory of aging.  相似文献   

5.
Animals search for foods and decide their behaviors according to previous experience. Caenorhabditis elegans detects chemicals with a limited number of sensory neurons, allowing us to dissect roles of each neuron for innate and learned behaviors. C. elegans is attracted to salt after exposure to the salt (NaCl) with food. In contrast, it learns to avoid the salt after exposure to the salt without food. In salt-attraction behavior, it is known that the ASE taste sensory neurons (ASEL and ASER) play a major role. However, little is known about mechanisms for learned salt avoidance. Here, through dissecting contributions of ASE neurons for salt chemotaxis, we show that both ASEL and ASER generate salt chemotaxis plasticity. In ASER, we have previously shown that the insulin/PI 3-kinase signaling acts for starvation-induced salt chemotaxis plasticity. This study shows that the PI 3-kinase signaling promotes aversive drive of ASER but not of ASEL. Furthermore, the Gq signaling pathway composed of Gqα EGL-30, diacylglycerol, and nPKC (novel protein kinase C) TTX-4 promotes attractive drive of ASER but not of ASEL. A putative salt receptor GCY-22 guanylyl cyclase is required in ASER for both salt attraction and avoidance. Our results suggest that ASEL and ASER use distinct molecular mechanisms to regulate salt chemotaxis plasticity.ANIMALS show various behaviors in response to environmental cues and modulate behaviors according to previous experience. To understand neuronal plasticity underlying learning, it is important to dissect neurons and molecules for sensing environmental stimuli, storing memory, and executing learned behaviors.The nematode Caenorhabditis elegans has only 302 neurons and functions of sensory neurons are well characterized (White et al. 1986; Bargmann 2006). C. elegans is attracted to odorants sensed by the AWC olfactory neurons or to salts sensed by the ASE gustatory neurons (Bargmann and Horvitz 1991; Bargmann et al. 1993). The ASE neuron class consists of a bilaterally symmetrical pair, ASE-left (ASEL) and ASE-right (ASER), which sense different sets of ions including Na+ and Cl, respectively (Pierce-Shimomura et al. 2001; Suzuki et al. 2008; Ortiz et al. 2009). ASEL is activated by an increase in salt concentration, whereas ASER is activated by a decrease in salt concentration (Suzuki et al. 2008). In the ASE gustatory neurons, a cyclic GMP (cGMP) signaling pathway mediates sensory transduction (Komatsu et al. 1996; Suzuki et al. 2008; Ortiz et al. 2009). ASEL and ASER express different sets of receptor-type guanylyl cyclases (gcys) (Ortiz et al. 2006). Of these, gcy-22, which is specifically expressed in ASER, is important for attraction to ASER-sensed ions such as Cl (Ortiz et al. 2009).Preference for salts changes according to previous experience (known as gustatory plasticity or salt chemotaxis learning) (Saeki et al. 2001; Jansen et al. 2002; Tomioka et al. 2006). When worms are grown on a medium that contains sodium chloride (NaCl) and food (Escherichia coli), they show attraction to NaCl by using ASE neurons (Bargmann and Horvitz 1991; Suzuki et al. 2008). In contrast, after exposure to the salt under starvation conditions, they show reduced attraction to or even avoid the salt (Saeki et al. 2001; Jansen et al. 2002; Tomioka et al. 2006). In C. elegans, it was proposed that preference for a sensory cue is defined by the sensory neuron that detects the cue (Troemel et al. 1997). ASE neurons play a major role for salt attraction (Bargmann and Horvitz 1991; Suzuki et al. 2008; Ortiz et al. 2009). However, little is known about sensory neurons that drive the learned salt avoidance; it remains unclear whether ASE neurons act as salt receptors for the learned avoidance.We have previously shown that an insulin/PI 3-kinase signaling pathway is essential for salt chemotaxis learning (Tomioka et al. 2006). In C. elegans, the insulin-like signaling is composed of daf-2, age-1, and akt-1, which encode homologs of insulin receptor, PI 3-kinase, and protein kinase B, respectively (Morris et al. 1996; Kimura et al. 1997; Paradis and Ruvkun 1998). Mutants of daf-2, age-1, and akt-1 show attraction to salt even after starvation/NaCl conditioning (Tomioka et al. 2006).daf-18 encodes a homolog of phosphatase PTEN (phosphatase and tensin homolog deleted on chromosome ten), which dephosphorylates phosphatidylinositol (3,4,5)-triphosphate and counteracts the insulin/PI 3-kinase signaling (Ogg and Ruvkun 1998; Gil et al. 1999; Mihaylova et al. 1999; Rouault et al. 1999; Solari et al. 2005). Mutants of daf-18, in which the PI 3-kinase signaling is activated, show reduced attraction to NaCl even without conditioning. Since the insulin/PI 3-kinase signaling acts in ASER, we proposed that the insulin/PI 3-kinase signaling attenuates the attractive drive of ASER (Tomioka et al. 2006).In C. elegans, diacylglycerol (DAG) regulates functions of motor neurons and sensory neurons. egl-30, which encodes the α-subunit of heterotrimeric G-protein Gq, facilitates production of DAG and enhances locomotory movements (Brundage et al. 1996; Lackner et al. 1999). In the AWC olfactory neurons, a novel protein kinase C-ɛ/η (nPKC-ɛ/η) ortholog TTX-4 (also known as PKC-1), which is one of DAG targets, plays an essential role in attraction behavior to AWC-sensed odors (Okochi et al. 2005; Tsunozaki et al. 2008). GOA-1 Goα regulates olfactory adaptation by antagonizing Gqα–DAG signaling (Matsuki et al. 2006).This study investigated the involvement of the ASE taste receptor neurons in the starvation-induced salt avoidance. We show that both ASEL and ASER contribute to salt chemotaxis learning. Activation of the PI 3-kinase signaling and the Gq/DAG/PKC signaling acted antagonistically in reversal of ASER function, whereas these signaling pathways did not have prominent effects on ASEL function. In ASER, GCY-22 was required for both salt attraction and avoidance. These results suggest that ASE neurons are important for bidirectional chemotaxis and also suggest that distinct molecular mechanisms regulate functions of ASEL and ASER in salt chemotaxis learning.  相似文献   

6.
The components of receptor tyrosine kinase signaling complexes help to define the specificity of the effects of their activation. The Caenorhabditis elegans fibroblast growth factor receptor (FGFR), EGL-15, regulates a number of processes, including sex myoblast (SM) migration guidance and fluid homeostasis, both of which require a Grb2/Sos/Ras cassette of signaling components. Here we show that SEM-5/Grb2 can bind directly to EGL-15 to mediate SM chemoattraction. A yeast two-hybrid screen identified SEM-5 as able to interact with the carboxy-terminal domain (CTD) of EGL-15, a domain that is specifically required for SM chemoattraction. This interaction requires the SEM-5 SH2-binding motifs present in the CTD (Y1009 and Y1087), and these sites are required for the CTD role of EGL-15 in SM chemoattraction. SEM-5, but not the SEM-5 binding sites located in the CTD, is required for the fluid homeostasis function of EGL-15, indicating that SEM-5 can link to EGL-15 through an alternative mechanism. The multi-substrate adaptor protein FRS2 serves to link vertebrate FGFRs to Grb2. In C. elegans, an FRS2-like gene, rog-1, functions upstream of a Ras/MAPK pathway for oocyte maturation but is not required for EGL-15 function. Thus, unlike the vertebrate FGFRs, which require the multi-substrate adaptor FRS2 to recruit Grb2, EGL-15 can recruit SEM-5/Grb2 directly.FIBROBLAST growth factors (FGFs) play important roles in many developmental and physiological processes, including cell migration, angiogenesis, proliferation, differentiation, and survival (Ornitz and Itoh 2001; Polanska et al. 2009). Mammals have a battery of both FGF ligands and high-affinity receptors to carry out this diverse set of important functions. These ligands and their receptors are generated from a set of 18 genes encoding FGFs and 4 genes encoding their receptors (Eswarakumar et al. 2005). Upon ligand binding, fibroblast growth factor receptors (FGFRs) dimerize, activating their intrinsic tyrosine kinase activity, which causes both autophosphorylation on intracellular tyrosine residues and phosphorylation of additional substrates (Eswarakumar et al. 2005). These phosphorylation events lead to the assembly of a signaling complex around the activated receptor, ultimately promoting various downstream signaling pathways (Eswarakumar et al. 2005).A large portion of mammalian FGFR signaling is mediated by the multi-substrate adaptor protein FRS2/snt-1 (Kouhara et al. 1997; Hadari et al. 1998, 2001; Lax et al. 2002; Gotoh et al. 2005). FRS2 constitutively associates with the juxtamembrane region of the FGFR via its amino-terminal PTB domain (Xu et al. 1998; Ong et al. 2000). Upon FGFR activation, FRS2 becomes heavily phosphorylated, allowing it to recruit Grb2 and Shp2 via their SH2 domains (Kouhara et al. 1997; Eswarakumar et al. 2005). Since these components cannot associate with the receptor in the absence of FRS2 (Hadari et al. 2001), FRS2 serves as an essential link between the activated receptor and many downstream signal transduction pathways.The understanding of FGF-stimulated signal transduction pathways has been aided by the study of FGF signaling in model organisms. Powerful genetic screens and the reduced complexity of the set of FGFs and their receptors in both Drosophila melanogaster and Caenorhabditis elegans have helped promote an understanding of the conserved aspects of FGF signaling pathways (Huang and Stern 2005; Polanska et al. 2009). In C. elegans, FGF signaling is mediated by two FGF ligands, EGL-17 and LET-756, and a single FGF receptor, EGL-15 (DeVore et al. 1995; Burdine et al. 1997; Roubin et al. 1999). The EGL-15 FGFR is structurally very similar to mammalian FGF receptors, with the highest level of sequence conservation found within the intracellular tyrosine kinase domain and the three extracellular immunoglobulin (IG) domains.Similar to mammalian FGFRs, alternative splicing also generates functionally distinct EGL-15 isoforms. A major structural difference between EGL-15 and other FGFRs lies in an additional domain located between the first IG domain and the acid box of EGL-15. This EGL-15-specific insert is encoded by a pair of mutually exclusive fifth exons, generating two EGL-15 isoforms, 5A and 5B, with different functions (Goodman et al. 2003). Alternative splicing also affects the sequence at the very end of the carboxy-terminal domain (CTD) of EGL-15 (Goodman et al. 2003), giving rise to four distinct C-terminal isoform types, referred to as types I–IV (see Figure 1A).Open in a separate windowFigure 1.—The SEM-5∷EGL-15 interaction is dependent upon the SEM-5 SH2 binding sites in the CTD of EGL-15. (A) Important residues in the EGL-15 CTD isoforms. The C-terminal portion of the kinase domain and the CTD are shown. The gray box is common to all C-terminal isoforms. Sequences of the CTD isoforms can be found in Figure S5. (B) SEM-5 binds directly to EGL-15 Y1009 and Y1087. A full-length SEM-5 prey was mated to eight EGL-15 baits: empty vector control (pBTM116) and derivatives containing variants of either the type I or the type IV EGL-15(Intra). Growth is an indication of an interaction between SEM-5 and the EGL-15 bait. Dilutions of the culture mixture are indicated above. hFGFR1, a bait containing the corresponding portion of the intracellular domain of the human FGFR1. Similar results were obtained using the human SEM-5 ortholog, Grb2.EGL-15, like its mammalian orthologs, is also involved in a large variety of functions, including cell migration guidance affecting the sex myoblast (SM) (Stern and Horvitz 1991; DeVore et al. 1995; Goodman et al. 2003) and CAN cells (Fleming et al. 2005), muscle arm extension (Dixon et al. 2006), a number of processes controlling terminal axon morphology (Bulow et al. 2004), muscle protein degradation (Szewczyk and Jacobson 2003), and fluid homeostasis (Huang and Stern 2004). A conserved FGFR signaling pathway in C. elegans was established by identifying the genes necessary for the role of EGL-15 in fluid homeostasis, and much of this same pathway is utilized in other functions of EGL-15 (DeVore et al. 1995; Borland et al. 2001; Huang and Stern 2004, 2005). The identification of these genes was facilitated by a temperature-sensitive mutation affecting a receptor tyrosine phosphatase, CLR-1 (CLeaR), which functions to negatively regulate EGL-15 signaling (Kokel et al. 1998). Mutations in clr-1 abolish this regulatory constraint on EGL-15, resulting in fluid accumulation in the pseudocoelomic cavity due to hyperactive EGL-15 signaling. The buildup of this clear fluid, resulting in the Clr phenotype, is easily scored and can be used to identify suppressors (soc, suppressor of Clr) that reduce EGL-15 signaling efficiency. These suppressors define an EGL-15 signaling pathway necessary for fluid homeostasis, which involves the activation of the Ras/MAPK cascade via the SEM-5/Grb2 adaptor protein, the let-341/sos-1 Sos-like guanine nucleotide exchange factor, and a PTP-2-SOC-1/Shp2-Gab1 cassette (Borland et al. 2001).Mutations in egl-15 have been identified on the basis of their effects on either fluid homeostasis or the guidance of the migrating SMs (DeVore et al. 1995; Goodman et al. 2003). Most of these alleles fall into an allelic series and affect the general aspects of EGL-15 signaling (Goodman et al. 2003). The strongest of these alleles confers an early larval arrest phenotype, whereas weaker alleles confer either a scrawny body morphology (Scr) or just the ability to suppress the Clr phenotype (Soc). While the weakest of these alleles does not affect egg laying, more highly compromised mutants show an egg-laying defect due to the mispositioning of the SMs. Four egl-15 alleles specifically affect SM migration; when homozygous, these mutations cause dramatic mispositioning of the SMs, but do not cause a Soc phenotype (Goodman et al. 2003). Three of these are nonsense mutations in exon 5A and eliminate the 5A EGL-15 isoform. The phenotype of these mutants highlights the specific requirement of the 5A isoform for SM migration guidance. The fourth mutation in this class, egl-15(n1457), is a nonsense mutation that truncates the carboxy-terminal domain, specifically implicating the CTD in SM migration guidance.Immediately following their birth at the end of the first larval stage (L1), the two bilaterally symmetric SMs undergo anteriorly directed migrations to final positions that flank the precise center of the gonad (Sulston and Horvitz 1977). In the middle of the third larval stage, the SMs divide to generate 16 cells that differentiate into the egg-laying muscles. Multiple mechanisms help guide the migrations of the SMs (Chen and Stern 1998; Branda and Stern 2000), including a chemoattraction mediated by EGL-15 that guides the SMs to their precise final positions (Burdine et al. 1998). The EGL-17 FGF serves as the chemoattractive cue, emanating from central gonadal cells (Branda and Stern 2000). In the absence of this chemoattraction, SMs are posteriorly displaced (Stern and Horvitz 1991). While mispositioned SMs still generate sex muscles, these muscles end up too far posterior to attach properly, causing the animal to be defective in egg laying (Egl).The signal transduction pathway downstream of EGL-15 that mediates SM chemoattraction is not well established. Several lines of evidence implicate SEM-5/Grb2, LET-341/Sos, and LET-60/Ras in SM chemoattraction. The roles of components in the Ras-MAPK cascade in this event are less clear (Sundaram et al. 1996; Chen et al. 1997; Chen and Stern 1998). A crucial gap in our understanding lies in the link between activated EGL-15 and the downstream signaling components. Here we show that SEM-5, the C. elegans GRB2 ortholog, appears to bind directly to SH2 binding sites within the carboxy terminal tail of EGL-15. These interactions are required for SM chemoattraction, but not for the essential function of EGL-15.  相似文献   

7.
In the developing nervous system, cohorts of events regulate the precise patterning of axons and formation of synapses between presynaptic neurons and their targets. The conserved PHR proteins play important roles in many aspects of axon and synapse development from C. elegans to mammals. The PHR proteins act as E3 ubiquitin ligases for the dual-leucine-zipper-bearing MAP kinase kinase kinase (DLK MAPKKK) to regulate the signal transduction cascade. In C. elegans, loss-of-function of the PHR protein RPM-1 (Regulator of Presynaptic Morphology-1) results in fewer synapses, disorganized presynaptic architecture, and axon overextension. Inactivation of the DLK-1 pathway suppresses these defects. By characterizing additional genetic suppressors of rpm-1, we present here a new member of the DLK-1 pathway, UEV-3, an E2 ubiquitin-conjugating enzyme variant. We show that uev-3 acts cell autonomously in neurons, despite its ubiquitous expression. Our genetic epistasis analysis supports a conclusion that uev-3 acts downstream of the MAPKK mkk-4 and upstream of the MAPKAPK mak-2. UEV-3 can interact with the p38 MAPK PMK-3. We postulate that UEV-3 may provide additional specificity in the DLK-1 pathway by contributing to activation of PMK-3 or limiting the substrates accessible to PMK-3.CHEMICAL synapses are specialized cellular junctions that enable neurons to communicate with their targets. An electrical impulse causes calcium channel opening and consequently stimulates synaptic vesicles in the presynaptic terminals to fuse at the plasma membrane. Neurotransmitter activates receptors on the postsynaptic membrane and triggers signal transduction in the target cell. For this communication to occur efficiently, the organization of the proteins within these juxtaposed pre- and postsynaptic terminals must be tightly regulated (Jin and Garner 2008). Previous studies in Caenorhabditis elegans have identified RPM-1, a member of the conserved PHR (Pam/Highwire/RPM-1) family of proteins, as an important regulator for the synapse (Schaefer et al. 2000; Zhen et al. 2000). Recent functional studies of other PHR proteins have shown that they are also required for a number of steps during nervous system development including axon guidance, growth, and termination (Wan et al. 2000; D''souza; et al. 2005; Bloom et al. 2007; Grill et al. 2007; Lewcock et al. 2007; Li et al. 2008).The signaling cascades regulated by the PHR proteins have been identified using genetic modifier screens (Diantonio et al. 2001; Liao et al. 2004; Nakata et al. 2005; Collins et al. 2006) and biochemical approaches (Grill et al. 2007; Wu et al. 2007). These studies reveal that a major function of PHR proteins is to act as ubiquitin E3 ligases (Jin and Garner 2008). In C. elegans, RPM-1 (Regulator of Presynaptic Morphology-1) regulates the abundance of its substrate, the dual-leucine-zipper-bearing MAP kinase kinase kinase (DLK MAPKKK), and controls the activity of the MAP kinase cascade composed of three additional kinases, MAPKK MKK-4, p38 MAPK PMK-3, and MAPKAPK MAK-2 (Nakata et al. 2005; Yan et al. 2009). This signaling cascade further regulates the activity of the CCAAT/enhancer binding protein (C/EBP), CEBP-1, via a mechanism involving 3′-UTR-mediated mRNA decay.Signal transduction involving MAP kinases can be fine tuned using multiple mechanisms to ensure optimal signaling outputs (Raman et al. 2007). For example, scaffold proteins for MAP kinases can provide spatial regulation of kinase activation in response to different stimuli (Remy and Michnick 2004; Whitmarsh 2006). Small protein tags such as ubiquitin have also been shown to control the activation of kinases. Specifically, in the IKK pathway ubiquitination via Lys63 chain formation catalyzed by the Ubc13/Uev1a E2 complex and TRAF6 E3 ligase is required for TAK1 kinase activation (Skaug et al. 2009).To further the understanding of the DLK-1 pathway in the development of the nervous system, we characterized a new complementation group of rpm-1(lf) suppressors. These mutations affect the gene uev-3, a ubiquitin E2 conjugating (UBC) enzyme variant (UEV). UEV proteins belong to the UBC family, but lack the catalytic active cysteine necessary for conjugating ubiquitin (Sancho et al. 1998). The best characterized UEV proteins are yeast Mms2 and mammalian Uev1A, both of which act as the obligatory partner for the active E2 Ubc13 and function in DNA repair and IKB pathways, respectively (Deng et al. 2000; Hurley et al. 2006). In addition, UEV proteins, such as Tsg101, can also regulate endosomal trafficking (Babst et al. 2000). We find that similar to other members of the DLK-1 pathway, uev-3 functions cell autonomously in neurons. uev-3 genetically acts downstream of mkk-4 and upstream of mak-2. UEV-3 can bind PMK-3 in heterologous protein interaction assays. We hypothesize that UEV-3 may add specificity to the DLK-1 pathway by binding to PMK-3 for its activation or for selecting specific downstream targets.  相似文献   

8.
9.
Primary cilia have essential roles in transducing signals in eukaryotes. At their core is the ciliary axoneme, a microtubule-based structure that defines cilium morphology and provides a substrate for intraflagellar transport. However, the extent to which axonemal microtubules are specialized for sensory cilium function is unknown. In the nematode Caenorhabditis elegans, primary cilia are present at the dendritic ends of most sensory neurons, where they provide a specialized environment for the transduction of particular stimuli. Here, we find that three tubulin isotypes—the α-tubulins TBA-6 and TBA-9 and the β-tubulin TBB-4—are specifically expressed in overlapping sets of C. elegans sensory neurons and localize to the sensory cilia of these cells. Although cilia still form in mutants lacking tba-6, tba-9, and tbb-4, ciliary function is often compromised: these mutants exhibit a variety of sensory deficits as well as the mislocalization of signaling components. In at least one case, that of the CEM cephalic sensory neurons, cilium architecture is disrupted in mutants lacking specific ciliary tubulins. While there is likely to be some functional redundancy among C. elegans tubulin genes, our results indicate that specific tubulins optimize the functional properties of C. elegans sensory cilia.THE fitness of all organisms depends on an ability to appropriately sense and respond to the environment. At the cellular level, many specific architectures have evolved to optimize these sensory functions. Prominent among these is the sensory cilium, a tubulin-based cytoplasmic extension that interrogates the extracellular environment in many biological contexts (Davenport and Yoder 2005; Berbari et al. 2009). Cilia are important for the transduction of a broad range of visual, auditory, mechanical, thermal, and chemical stimuli. They also function during development to receive a variety of signals, both chemical and mechanical, that regulate proliferation and differentiation (Goetz and Anderson 2010). Indeed, the disruption of cilium assembly and function can give rise to a spectrum of human diseases collectively known as ciliopathies (Berbari et al. 2009; Lancaster and Gleeson 2009). These disorders, which include autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD), Bardet–Biedl syndrome, Meckel–Gruber syndrome, and Joubert syndrome, are associated with a variety of pathogenic conditions including polycystic kidneys and neurological impairments.At the core of all cilia and flagella is the microtubule axoneme. This characteristic structural element comprises nine doublet outer microtubules that may surround a central pair, the presence of which often indicates a motile cilium/flagellum. Like all microtubule-based structures, ciliary axonemes are built of heterodimers of α- and β-tubulins, highly conserved small GTP-binding proteins. The recruitment of other cilium components, including signal transduction machinery, requires a conserved assembly and maintenance process called intraflagellar transport (IFT) (Blacque et al. 2008; Pedersen and Rosenbaum 2008). IFT employs two major complexes that transport ciliary cargo bidirectionally by traveling along the axonemal microtubules. Loss of individual IFT components can cause a broad spectrum of defects in the assembly, maintenance, and function of cilia.Important insights into cilium structure and function have come from studies of genetically tractable organisms, particularly the green alga Chlamydomonas and the nematode Caenorhabditis elegans (Bae and Barr 2008; Pedersen and Rosenbaum 2008). In C. elegans, sensory cilia are found exclusively at the dendritic ends of sensory neurons. These cilia constitute a highly specialized sensory environment characterized by localized sensory receptors and specific signaling components. Cilium morphology is quite distinctive in many of these cells and likely contributes to their functional specialization (Ward et al. 1975). Recent progress has shed light on the mechanisms that confer this specialization onto more general pan-ciliary pathways (Evans et al. 2006; Mukhopadhyay et al. 2007; Jauregui et al. 2008; Mukhopadhyay et al. 2008; Silverman and Leroux 2009).The genomes of many eukaryotes harbor multiple α- and β-tubulin genes. Two hypotheses, which are not mutually exclusive, have been proposed to account for these paralogs (Cleveland 1987; Wade 2007). At one extreme, different tubulin isotypes might be functionally redundant, such that their minor coding differences are largely irrelevant. According to this model, multiple genes allow the maintenance of a stable pool of available monomers and dimers. The small amount of sequence variation within the α- and β-tubulin families supports this idea, as do studies of functionally redundant mitotic tubulins in C. elegans (Ellis et al. 2004; Lu et al. 2004; Phillips et al. 2004; Lu and Mains 2005). The alternative hypothesis proposes that specific structures, e.g., ciliary axonemes or axonal microtubules, rely on tubulins optimized for specific roles. Support for this idea has come from studies of cultured mammalian neurons (Joshi and Cleveland 1989), Drosophila (Hutchens et al. 1997; Raff et al. 1997), and human tubulins (Vent et al. 2005; Jaglin et al. 2009). In Drosophila, studies of motile sperm flagella have revealed that the sperm-specific β2 tubulin isoform builds not only the specialized motile axoneme but also all other tubulin-based structures (Kemphues et al. 1982). However, sequences both within and outside the axoneme motif in the C-terminal tail of this tubulin isoform are required for the flagellar axoneme, and other closely related β-tubulins cannot support this role (Fuller et al. 1987; Raff et al. 1997; Popodi et al. 2008). Genetic interactions have provided evidence that β2 tubulin heterodimerizes with the α-tubulin 84B (Hays et al. 1989), which also possesses specific functional properties not provided by structurally similar α-tubulins (Hutchens et al. 1997). In C. elegans, a specific role for tubulin isoforms has been described in the six touch receptor neurons. These nonciliated cells harbor unusual 15-filament microtubules composed of dimers of the α-tubulin MEC-12 and the β-tubulin MEC-7. The loss of mec-7 or mec-12, the expression of which is largely restricted to these cells, results in the conversion of 15-filament microtubules to the standard 11-microfilament variety and a commensurate loss of light-touch response (Savage et al. 1989; Fukushige et al. 1999; Bounoutas et al. 2009). Thus experimental support exists for both of these opposing views, and it seems likely that the role of specific tubulin isoforms in regulating microtubule structure and function differs according to cell and organelle type.The C. elegans genome encodes nine α- and six β-tubulin genes (Gogonea et al. 1999). Some of these genes, particularly tba-1, tba-2, tbb-1, and tbb-2, are expressed broadly during embryogenesis and function redundantly in spindle assembly and positioning (Ellis et al. 2004; Lu et al. 2004; Phillips et al. 2004; Lu and Mains 2005). tba-1 and tbb-2 have also been recently shown to be important for axon outgrowth and synaptogenesis (Baran et al. 2010). Several others, including mec-7, mec-12, and the β-tubulin ben-1, have been identified through genetic screens for particular phenotypes, such as touch insensitivity or benzimidazole resistance (Driscoll et al. 1989; Savage et al. 1989; Fukushige et al. 1999). However, the extent to which specific tubulin isoforms are required for structural and functional diversity in the C. elegans nervous system remains unknown. Here, taking advantage of several existing genome-wide data sets, we identify the α-tubulins TBA-6 and TBA-9 and the β-tubulin TBB-4 as strong candidates for tubulins that have roles in sensory cilia. We find that each of these genes are expressed in characteristic, partially overlapping, sets of sensory neurons, where their products localize to ciliary axonemes. While the loss of any one (or all three) of these genes does not abolish ciliogenesis, tubulin mutants exhibit significant defects in the localization of cilium proteins and in some cilium-dependent behavioral responses. Together, our results indicate that specific α- and β-tubulin isoforms are important, although not essential, for the efficient assembly and function of specific classes of C. elegans sensory cilia. Sensory cilia throughout the animal kingdom may therefore employ specific tubulin isoforms to optimize their function.  相似文献   

10.
Fluoxetine is one of the most commonly prescribed medications for many behavioral and neurological disorders. Fluoxetine acts primarily as an inhibitor of the serotonin reuptake transporter (SERT) to block the removal of serotonin from the synaptic cleft, thereby enhancing serotonin signals. While the effects of fluoxetine on behavior are firmly established, debate is ongoing whether inhibition of serotonin reuptake is a sufficient explanation for its therapeutic action. Here, we provide evidence of two additional aspects of fluoxetine action through genetic analyses in Caenorhabditis elegans. We show that fluoxetine treatment and null mutation in the sole SERT gene mod-5 eliminate serotonin in specific neurons. These neurons do not synthesize serotonin but import extracellular serotonin via MOD-5/SERT. Furthermore, we show that fluoxetine acts independently of MOD-5/SERT to regulate discrete properties of acetylcholine (Ach), gamma-aminobutyric acid (GABA), and glutamate neurotransmission in the locomotory circuit. We identified that two G-protein–coupled 5-HT receptors, SER-7 and SER-5, antagonistically regulate the effects of fluoxetine and that fluoxetine binds to SER-7. Epistatic analyses suggest that SER-7 and SER-5 act upstream of AMPA receptor GLR-1 signaling. Our work provides genetic evidence that fluoxetine may influence neuronal functions and behavior by directly targeting serotonin receptors.FLUOXETINE is a selective serotonin reuptake inhibitor (SSRI) and has made a major impact on the treatment of many behavioral disorders. The empirical action of SSRIs is blocking the serotonin reuptake transporter (SERT). SERT is localized in the plasma membrane and transports extracellular serotonin (5-HT) into the cytoplasm (Blakely et al. 1991; Hoffman et al. 1991), this being the major mechanism of terminating 5-HT signaling. Consequently, SSRIs are thought to exert therapeutic effects by blocking SERT from removal of 5-HT in the synaptic clef, thereby increasing the level of 5-HT signals (Schatzberg and Nemeroff 2004). However, several observations point to additional actions of SSRIs on the 5-HT system and neuronal functions. First, knockout of SERT in mouse caused a marked reduction of 5-HT in the brain (Bengel et al. 1998). Second, a variety of studies with cultured mammalian cells and mouse brain slices showed that SSRIs and tricyclic antidepressant agents (TCAs) have high affinities to many 5-HT receptor subtypes and act as agonists or antagonists depending on particular receptors being tested (Ni and Miledi 1997; Kroeze and Roth 1998; Eisensamer et al. 2003). Third, genetic analyses of the nematode Caenorhabditis elegans in our laboratory and others showed that fluoxetine and the TCA imipramine (Tofrani) could influence behavior independent of SERT function (Weinshenker et al. 1995; Ranganathan et al. 2001; Dempsey et al. 2005). In this study, we carried out a systematic survey of SSRIs treatment in C. elegans to gain new insights into actions of SSRIs on the 5-HT system and other neurotransmitter systems.In both vertebrates and invertebrates, 5-HT functions as a neuromodulator to either facilitate or inhibit synaptic transmission of other neurotransmitters (Fink and Gothert 2007). Modulation of synaptic activity by 5-HT signaling underscores the synaptic plasticity involved in stress responses, learning, adaptation, and memory (Kandel 2001; Zhang et al. 2005). The role of 5-HT in C. elegans was initially identified through pharmacological experiments showing that exogenous 5-HT can promptly induce changes in a variety of behaviors, including feeding, egg laying, and locomotion (Avery and Horvitz 1990; Weinshenker et al. 1995; Nurrish et al. 1999). The relevance of these behaviors to endogenous 5-HT has since been validated through studies of mutants of 5-HT signaling. Importantly, multiple 5-HT receptors may function in distinct cells synergistically or antagonistically to regulate a specific behavior (Carnell et al. 2005; Dernovici et al. 2007; Murakami and Murakami 2007; Hapiak et al. 2009). In nearly all tested paradigms, fluoxetine and imipramine induce behavioral changes similarly to exogenous 5-HT (Weinshenker et al. 1995; Nurrish et al. 1999), implying that fluoxetine regulates 5-HT inputs to these neural circuits. However, the tryptophan hydroxylase gene tph-1 is required for 5-HT biosynthesis in C. elegans (Sze et al. 2000), mod-5 encodes its sole SERT (Ranganathan et al. 2001), and yet fluoxetine could stimulate egg laying and inhibit locomotion in mod-5 and tph-1 mutants (Weinshenker et al. 1995; Choy and Thomas 1999; Ranganathan et al. 2001; Dempsey et al. 2005). These findings provided a basis for further investigation into genes and synaptic functions regulated by 5-HT and the impact of fluoxetine on 5-HT signaling.Here we present genetic evidence of multifaceted effects of fluoxetine on the 5-HT system and its downstream targets in C. elegans. We show that fluoxetine treatment and loss of MOD-5/SERT function do not simply increase presynaptic 5-HT signals. Rather, they may eliminate 5-HT in specific neurons. Furthermore, fluoxetine acts independently of SERT to regulate 5-HT serotonin receptors and their downstream targets involved in acetylcholine (ACh), gamma-aminobutyric acid (GABA), and glutamate neurotransmission.  相似文献   

11.
The polarization of post-mitotic neurons is poorly understood. Preexisting spatially asymmetric cues, distributed within the neuron or as extracellular gradients, could be required for neurons to polarize. Alternatively, neurons might have the intrinsic ability to polarize without any preestablished asymmetric cues. In Caenorhabditis elegans, the UNC-40 (DCC) receptor mediates responses to the extracellular UNC-6 (netrin) guidance cue. For the HSN neuron, an UNC-6 ventral-dorsal gradient asymmetrically localizes UNC-40 to the ventral HSN surface. There an axon forms, which is ventrally directed by UNC-6. In the absence of UNC-6, UNC-40 is equally distributed and the HSN axon travels anteriorly in response to other cues. However, we find that a single amino acid change in the UNC-40 ectodomain causes randomly oriented asymmetric UNC-40 localization and a wandering axon phenotype. With UNC-6, there is normal UNC-40 localization and axon migration. A single UNC-6 amino acid substitution enhances the mutant phenotypes, whereas UNC-6 second-site amino acid substitutions suppress the phenotypes. We propose that UNC-40 mediates multiple signals to polarize and orient asymmetry. One signal triggers the intrinsic ability of HSN to polarize and causes randomly oriented asymmetry. Concurrently, another signal biases the orientation of the asymmetry relative to the UNC-6 gradient. The UNC-40 ectodomain mutation activates the polarization signal, whereas different forms of the UNC-6 ligand produce UNC-40 conformational changes that allow or prohibit the orientation signal.A major challenge for developmental neuroscience has been to understand how axons are able to detect and follow molecular gradients of different extracellular guidance cues. Attractive guidance cues are proposed to stimulate cytoplasmic signaling pathways that promote actin polymerization (Huber et al. 2003). Thus the direction of axon outgrowth is directly linked to the extracellular gradient of the guidance cue; i.e., there is greater extension on the side of the neuron that is closest to the source of the cue. Netrins are bifunctional guidance cues that are attractive to some axons but repulsive to others. Studies have shown that the axon response to netrin is determined by the composition of netrin receptors on the cell surface and the internal state of the growth cone (Round and Stein 2007). The UNC-6 (netrin) guidance cue in Caenorhabditis elegans interacts with the UNC-40 (DCC) receptor to mediate attraction (Hedgecock et al. 1990; Ishii et al. 1992; Chan et al. 1996). The AVM and HSN neurons are useful for studying UNC-40-mediated responses to UNC-6. The cell bodies of these neurons are situated on the lateral body wall and send a single axon ventrally during larval development.In AVM and HSN, a signaling module comprising UNC-6, UNC-40, phosphoinositide 3-kinase (PI3K), Rac, and MIG-10 (lamellipodin) is thought to transmit the directional information provided by the graded distribution of extracellular guidance cues to the internal cellular machinery that promotes directed outgrowth (Adler et al. 2006; Chang et al. 2006; Quinn et al. 2006, 2008). MIG-10 appears to provide an important link because this family of proteins can interact with proteins that promote actin polymerization, and it is associated with asymmetric concentrations of f-actin and microtubules in turning growth cones (Krause et al. 2004; Quinn et al. 2008). MIG-10 is observed as asymmetrically localized to the ventral site of axon outgrowth in developing HSN neurons. This MIG-10 localization is sensitive to the source of UNC-6. Normally, the source of UNC-6 is ventral; in the absence of UNC-6, there is an equal distribution of MIG-10 along the cell surface, whereas ectopic UNC-6 expression from dorsal muscles causes dorsal MIG-10 localization (Adler et al. 2006). The UNC-40 receptor is also asymmetrically localized in HSN, and this localization is also dependent on UNC-6 (Adler et al. 2006). UNC-40 signaling activates Rac GTPase, and MIG-10 interacts specifically with the activated Rac (Quinn et al. 2008). Therefore, the asymmetric activation of Rac through UNC-40 recruits asymmetric MIG-10 localization.By activating or directing components to the surface nearest the UNC-6 source, the asymmetric distribution of UNC-6 could polarize the neuron. However, an alternative idea is suggested from studies of chemotaxing cells. This model predicts that chemoattractant signaling involves two different elements: one that activates the intrinsic ability of cells to generate asymmetry and another that biases the orientation of the asymmetry (Wedlich-Soldner and Li 2003). The polarization signal does not depend on the spatial information provided by the chemoattractant gradient, whereas the orientation signal does. The asymmetric localization of the UNC-40 and MIG-10 signaling complex is suggestive of the segregation of signaling components into separate “front” and “rear” regions during chemotactic cell migration (Weiner 2002; Mortimer et al. 2008). It is hypothesized that this segregation is accomplished through short-range positive feedback mechanisms that promote the local production or recruitment of signaling molecules. In addition, a long-range inhibition mechanism globally increases the degradation of these molecules. Together such mechanisms could strongly amplify the asymmetric distribution of molecules needed for directed movement. This model has been put forth to explain why chemotactic cells polarize and move in a random direction when encountering a uniform chemoattractant concentration. Although the chemoattractant receptors may be uniformly stimulated across the surface of the cells, randomly oriented asymmetry can be established through these mechanisms.If the AVM and HSN neurons behave similarly to chemotactic cells, then uniformly stimulating UNC-40 receptors might similarly cause nonspecific asymmetric UNC-40 localization and axon migrations in varying directions. However, this is difficult to test in vivo. Unlike exposing chemotactic cells to a uniform concentration of a chemotractant in vitro, there is no reliable way to ensure that a neuron in vivo is exposed to a uniform concentration of UNC-6. The pseudocoelomic cavity of C. elegans is fluid filled, and UNC-6 expression patterns are spatially and temporally complex (Wadsworth et al. 1996). How the distribution of UNC-6 is affected by interactions with the extracellular matrix and cell surfaces is unknown.Using a genetic approach, we have found an UNC-40 mutation that triggers randomly oriented neuronal asymmetry. On the basis of the models proposed for chemotactic cells, we suggest that there is an UNC-6/UNC-40-mediated signal that specifically induces the neuron''s intrinsic ability to polarize. The UNC-40 mutation activates this signal; however, a second signal, which normally would concurrently orient asymmetry relative to the UNC-6 gradient, is not activated. Single amino acid changes within the UNC-6 ligand can enhance or suppress the randomly oriented asymmetry phenotype caused by the UNC-40 mutation. This suggests that specific UNC-40 conformations uncouple the activation of the different signals.  相似文献   

12.
The synaptonemal complex (SC) is a highly ordered proteinaceous structure that assembles at the interface between aligned homologous chromosomes during meiotic prophase. The SC has been demonstrated to function both in stabilization of homolog pairing and in promoting the formation of interhomolog crossovers (COs). How the SC provides these functions and whether it also plays a role in inhibiting CO formation has been a matter of debate. Here we provide new insight into assembly and function of the SC by investigating the consequences of reducing (but not eliminating) SYP-1, a major structural component of the SC central region, during meiosis in Caenorhabditis elegans. First, we find an increased incidence of double CO (DCO) meiotic products following partial depletion of SYP-1 by RNAi, indicating a role for SYP-1 in mechanisms that normally limit crossovers to one per homolog pair per meiosis. Second, syp-1 RNAi worms exhibit both a strong preference for COs to occur on the left half of the X chromosome and a significant bias for SYP-1 protein to be associated with the left half of the chromosome, implying that the SC functions locally in promoting COs. Distribution of SYP-1 on chromosomes in syp-1 RNAi germ cells provides strong corroboration for cooperative assembly of the SC central region and indicates that SYP-1 preferentially associates with X chromosomes when it is present in limiting quantities. Further, the observed biases in the distribution of both COs and SYP-1 protein support models in which synapsis initiates predominantly in the vicinity of pairing centers (PCs). However, discontinuities in SC structure and clear gaps between localized foci of PC-binding protein HIM-8 and X chromosome-associated SYP-1 stretches allow refinement of models for the role of PCs in promoting synapsis. Our data suggest that the CO landscape is shaped by a combination of three attributes of the SC central region: a CO-promoting activity that functions locally at CO sites, a cooperative assembly process that enables CO formation in regions distant from prominent sites of synapsis initiation, and CO-inhibitory role(s) that limit CO number.REDUCTION in ploidy during sexual reproduction depends on the ability to form pairwise associations between homologous chromosomes. The homolog pairing process typically culminates in an arrangement in which the homologs are aligned in parallel along their lengths, with a highly ordered proteinaceous structure known as the synaptonemal complex (SC) located at the interface between them. Further, in most organisms, pairwise associations between homologs are solidified through the formation of crossovers (COs) between their DNA molecules, a process that is completed within the context of the SC.The SC has long been recognized as a hallmark cytological feature of meiosis. It was discovered on the basis of its highly ordered structure and location at the interface between aligned chromosomes in electron microscopy images of nuclei at the pachytene stage of meiotic prophase (Moses 1956, 2006). Each of the homologs is associated with one of the two lateral elements (LEs) of the SC, which are composed of cohesin complexes and other meiosis-specific structural and regulatory proteins (reviewed in Mlynarczyk-Evans and Villeneuve 2010). The LEs are connected by a highly ordered latticework of transverse filaments, and often a pronounced central element, that comprise the central region of the SC. The protein components of the SC central region are very poorly conserved at the primary sequence level, but the major central region proteins identified from diverse species share in common extended regions of predicted coiled coil structure.The SC has been demonstrated to have at least two conserved functions in meiotic prophase. First, the SC serves to stabilize and maintain tight associations along the lengths of aligned homologs (reviewed in Mlynarczyk-Evans and Villeneuve 2010). This is true both in organisms in which SC assembly is coupled to formation of recombination intermediates (e.g., budding yeast, mouse, and Arabidopsis) and in organisms in which formation of SC between homologs can occur independently of recombination (e.g., Caenorhabditis elegans and Drosophila). Second, SC central region proteins play a role in promoting maturation of recombination intermediates into crossover products (reviewed in De Boer and Heyting 2006). How the SC functions to promote CO formation is not well understood. Moreover, whether the SC might also have additional functions that help to ensure a successful outcome of meiosis has been a matter of debate.In addition to its roles in stabilization of pairing and promoting CO formation, the SC has also been proposed to function in inhibiting CO formation (Egel 1978, 1995; Maguire 1988). This idea of the SC playing an inhibitory role in recombination dates almost as far back as the discovery of the SC itself. Finding a highly ordered structure with a zipper-like appearance extending along the length of each homolog pair naturally gave rise to speculation that it might play a role in the phenomenon of crossover interference, defined as the ability of a (nascent) CO to inhibit the formation of other COs nearby on the same chromosome pair (Muller 1916; Hillers 2004). It was variously proposed either that the SC might serve as a conduit of information along a chromosome pair (e.g., undergoing a distance-dependent “change in state” to inhibit COs) or that SC polymerization might itself confer CO inhibition (Egel 1978; Maguire 1988; Sym and Roeder 1994).Early analysis of the budding yeast mutants lacking Zip1, a major structural component of the SC central region, initially seemed to support the idea that the SC central region played a key role in CO interference, as zip1 mutants formed COs at 30–50% of wild-type levels and the residual COs did not display interference (Sym and Roeder 1994). However, these data were subsequently reinterpreted by postulating that the major interference-sensitive meiotic CO pathway is eliminated in the zip1 mutant and that the residual COs form by an alternative pathway that is not subject to interference (Zalevsky et al. 1999; de los Santos et al. 2003). According to this two-pathway view, the lack of interference in the zip1 mutant can be readily explained without invoking a role for Zip1 in the interference mechanism per se. Conversely, Page and Hawley found that Drosophila females expressing a mutant form of the fly SC central region protein C(3)G retained substantial interference between residual COs despite exhibiting incomplete synapsis, implying that complete SC formation was not required for CO interference (Page and Hawley 2001). In light of these and other findings (e.g., Borner et al. 2004; Fung et al. 2004), the idea that the SC might play a role in inhibiting CO formation fell from favor.In this study, we revisit a potential role for the SC central region in inhibiting CO formation, using the C. elegans experimental system. Several features make this an interesting system for investigating factors that promote and/or inhibit COs during meiosis. First, essentially all COs in C. elegans depend on conserved meiotic CO-promoting machinery (i.e., Msh4 and Msh5) and on SC central region proteins (SYP-1, -2, -3, and -4), so analysis is generally not complicated by residual COs forming by alternative pathways (Zalevsky et al. 1999; Kelly et al. 2000; MacQueen et al. 2002; Colaiacovo et al. 2003; Smolikov et al. 2007a, 2009). Second, C. elegans hermaphrodites exhibit robust CO control, with COs usually being limited to one per homolog pair per meiosis (Hillers and Villeneuve 2003; Nabeshima et al. 2004; Hammarlund et al. 2005). Consequently, circumstances that give rise to double crossover (DCO) meiotic products can be inferred to represent impairment of mechanisms that normally inhibit CO formation. Finally, COs are distributed nonuniformly along the lengths of the chromosomes, with each chromosome containing broad domains of relatively high CO frequency flanking a more central domain where CO frequency is low (Brenner 1974; Barnes et al. 1995; Rockman and Kruglyak 2009), providing an opportunity to evaluate how factors that promote and/or inhibit COs contribute to this landscape.Our strategy was to use RNAi to reduce the levels of wild-type SYP-1 protein without eliminating synapsis entirely and then to examine the effects on CO frequency and distribution. This approach indeed revealed a role for SC central region protein SYP-1 in mechanisms that normally limit the number of COs per homolog pair. Further, it also revealed a role for the SC central region in determining CO distribution, presumably by enabling formation of COs in chromosome regions distant from the dominant site of synapsis initiation. Finally, our experimental design also afforded us the opportunity to evaluate spatial distribution of the SC in the context of limiting amounts of a key central region component. This analysis provided additional insight into the process of SC assembly and the role of cis-acting meiotic pairing centers in this process.  相似文献   

13.
The Caenorhabditis elegans somatic gonad develops from a four-cell primordium into a mature organ that differs dramatically between the sexes in overall morphology (two arms in hermaphrodites and one in males) and in the cell types comprising it. Gonadal development in C. elegans is well studied, but regulation of sexual differentiation, especially later in gonadal development, remains poorly elucidated. To identify genes involved in this process, we performed a genome-wide RNAi screen using sex-specifically expressed gonadal GFP reporters. This screen identified several phenotypic classes, including ∼70 genes whose depletion feminized male gonadal cells. Among the genes required for male cell fate specification are Wnt/β-catenin pathway members, cell cycle regulators, and genes required for mitotic spindle function and cytokinesis. We find that a Wnt/β-catenin pathway independent of extracellular Wnt ligand is essential for asymmetric cell divisions and male differentiation during gonadal development in larvae. We also find that the cell cycle regulators cdk-1 and cyb-3 and the spindle/cytokinesis regulator zen-4 are required for Wnt/β-catenin pathway activity in the developing gonad. After sex is determined in the gonadal primordium the global sex determination pathway is dispensable for gonadal sexual fate, suggesting that male cell fates are promoted and maintained independently of the global pathway during this period.THE Caenorhabditis elegans gonad derives from a simple primordium of four cells that coalesces during embryogenesis and contains two somatic gonad precursors (SGPs), Z1 and Z4, flanking two germline precursors, Z2 and Z3 (Kimble and Hirsh 1979). The SGPs undergo very different developmental programs in each sex, involving sexually dimorphic cell lineages and migrations and sex-specific cellular differentiation. The result is a two-armed bilaterally symmetrical gonad in the adult hermaphrodite or a single-armed asymmetric gonad in the adult male. The high degree of sexual dimorphism of the mature organ and variety of cellular events that occur sex specifically during its development make the C. elegans gonad an outstanding model for sex-specific organogenesis.Development of the somatic gonad occurs in two phases. The early phase defines the gonadal axes and establishes the precursors of the major gonadal cell types. This takes place during the first larval stage (L1), beginning shortly after hatching with the first division of the SGPs. In both sexes SGP division is asymmetric in terms of both the sizes and the fates of the daughter cells, and establishes the proximal/distal axis of the gonad (Hirsh et al. 1976; Kimble and Hirsh 1979). The global sex determination pathway establishes the future sex of the gonad around the time of hatching (Klass et al. 1976; Nelson et al. 1978), and sexual dimorphism is already apparent when the SGPs divide: the size asymmetry of the SGP daughters is much more pronounced in males than hermaphrodites. In both sexes the asymmetry of the first SGP division requires a Wnt/β-catenin pathway. Mutations compromising this pathway cause a “symmetrical sisters” phenotype in which both daughters adopt the same fate (Miskowski et al. 2001; Siegfried and Kimble 2002; Phillips and Kimble 2009). Sex specificity is imposed on the SGPs by the global sex determining gene tra-1 (Hodgkin 1987) and the gonad-specific sex determining gene fkh-6 (Chang et al. 2004). These genes play opposing roles in SGP sex determination, with tra-1 feminizing and fkh-6 masculinizing the somatic gonad, and they also act redundantly to promote mitotic proliferation of the SGP lineage (Chang et al. 2004). SGP sex determination is linked to cell cycle progression by cyclin D, which is required to overcome repression of fkh-6 expression in the SGPs by E2F (Tilmann and Kimble 2005).The later phase of gonadal development involves the elongation of the gonad, together with cellular proliferation and differentiation, and lasts from L2 to adulthood. During L2 the somatic cells enlarge and leader cells (distal tip cells in the hermaphrodite, linker cell in the male) begin long-range migrations that extend the gonad. During L3, somatic gonad cell division resumes in both sexes, leading to the formation of differentiated somatic cell types by the end of L3 or beginning of L4. Gonadal morphogenesis is completed and gametogenesis begins during L4 (Kimble and Hirsh 1979).Although SGP division and much of hermaphrodite gonadal development have been well studied (Hubbard and Greenstein 2000), sexual cell fate specification in the somatic gonad is more poorly understood, particularly after the L1 stage. Despite the importance of fkh-6 in promoting male differentiation, it is expressed in males only during early L1 and null mutants have incomplete gonadal sex reversal. We have therefore performed a genome-wide RNAi screen to identify additional genes required after hatching for gonadal development in each sex. Among the advantages of this approach is the ability to identify gonadal regulators that also are essential for embryonic development. To our knowledge this is the first functional genomic study of gonadal sex differentiation.The screen identified many genes whose depletion disrupts gonadogenesis in each sex and nearly 70 genes whose depletion causes gonadal feminization in males. Prominent among this latter class were components of a Wnt/β-catenin pathway, cell cycle regulators, and genes involved in mitotic spindle function and cytokinesis. We find that Wnt/β-catenin activity continues in both sexes after SGP division and is required for male cell fate commitment in the gonad. We also find that the cyclin-dependent kinase cdk-1 and its cognate cyclin cyb-3 as well as the mitotic spindle regulator zen-4 are required for gonadal Wnt/β-catenin pathway activity, providing a potential new link between the cell cycle, asymmetric division, and sexual differentiation. The feminization caused by depletion of Wnt/β-catenin pathway components or cdk-1 is independent of the global sex determination pathway, suggesting that sexual fates in the male gonad remain plastic after the primary sex determination decision.  相似文献   

14.
15.
16.
Jae Hoon Bahn  Gyunghee Lee    Jae H. Park 《Genetics》2009,181(3):965-975
PAR proteins (partitioning defective) are major regulators of cell polarity and asymmetric cell division. One of the par genes, par-1, encodes a Ser/Thr kinase that is conserved from yeast to mammals. In Caenorhabditis elegans, par-1 governs asymmetric cell division by ensuring the polar distribution of cell fate determinants. However the precise mechanisms by which PAR-1 regulates asymmetric cell division in C. elegans remain to be elucidated. We performed a genomewide RNAi screen and identified six genes that specifically suppress the embryonic lethal phenotype associated with mutations in par-1. One of these suppressors is mpk-1, the C. elegans homolog of the conserved mitogen activated protein (MAP) kinase ERK. Loss of function of mpk-1 restored embryonic viability, asynchronous cell divisions, the asymmetric distribution of cell fate specification markers, and the distribution of PAR-1 protein in par-1 mutant embryos, indicating that this genetic interaction is functionally relevant for embryonic development. Furthermore, disrupting the function of other components of the MAPK signaling pathway resulted in suppression of par-1 embryonic lethality. Our data therefore indicates that MAP kinase signaling antagonizes PAR-1 signaling during early C. elegans embryonic polarization.ASYMMETRIC cell division, a process in which a mother cell divides in two different daughter cells, is a fundamental mechanism to achieve cell diversity during development. We use the early embryo of Caenorhabditis elegans as a model system to study asymmetric cell division. The C. elegans one-cell embryo divides asymmetrically along its anteroposterior axis, generating two cells of different sizes and fates: the larger anterior daughter cell will generate somatic tissues while the smaller posterior daughter cell will generate the germline (Sulston et al. 1983).A group of proteins called PAR proteins (partitioning defective) is required for asymmetric cell division in C. elegans (Kemphues et al. 1988). Depletion of any of the seven par genes (par-1 to -6 and pkc-3) leads to defects in asymmetric cell division and embryonic lethality (Kemphues et al. 1988; Kirby et al. 1990; Tabuse et al. 1998; Hung and Kemphues 1999; Hao et al. 2006). PAR-3 and PAR-6 are conserved proteins that contain PDZ-domains and form a complex with PKC-3 (Etemad-Moghadam et al. 1995; Izumi et al. 1998; Tabuse et al. 1998; Hung and Kemphues 1999). This complex becomes restricted to the anterior cortex of the embryo in response to spatially defined actomyosin contractions occurring in the embryo upon fertilization (Goldstein and Hird 1996; Munro et al. 2004). The posterior cortex of the embryo that becomes devoid of the anterior PAR proteins is occupied by the RING protein PAR-2 and the Ser/Thr kinase PAR-1 (Guo and Kemphues 1995; Boyd et al. 1996; Cuenca et al. 2003). Once polarized, the anterior and posterior PAR proteins mutually exclude each other from their respective cortices (Etemad-Moghadam et al. 1995; Boyd et al. 1996; Cuenca et al. 2003; Hao et al. 2006). Loss of function of the gene par-1, as opposed to loss of most other par genes, results in embryos that exhibit only subtle effects on the polarized cortical domains occupied by the other PAR proteins (Cuenca et al. 2003). However defects in this gene are associated with a more symmetric division in size, an aberrant distribution of cell fate specification markers, altered cell fates of the daughter cells of the embryo, and ultimately embryonic lethality (Kemphues et al. 1988; Guo and Kemphues 1995).PAR-1 controls asymmetric cell division and cell fate specification by regulating the localization of the two highly similar CCCH-type zinc-finger proteins MEX-5 and MEX-6 (referred to as MEX-5/6). MEX-5 and MEX-6 are 70% identical in their amino acid sequence and fulfill partially redundant functions in the embryo (Schubert et al. 2000). In wild-type animals, endogenous MEX-5 and GFP fusions of MEX-6 localize primarily to the anterior of the embryo while both proteins are evenly distributed in par-1 mutant embryos (Schubert et al. 2000; Cuenca et al. 2003). This suggests that in wild-type animals, PAR-1 acts in part by restricting MEX-5 and MEX-6 to the anterior of the embryo. The precise mechanism of this regulation is not known, but an elegant study performed for MEX-5 indicates that differential protein mobility in the anterior and posterior cytoplasm of the one-cell embryo contributes to this asymmetry (Tenlen et al. 2008). While increased mobility in the posterior of the one-cell embryo correlates with a par-1- and par-4-dependent phosphorylation on MEX-5, the kinase directly phosphorylating MEX-5 remains to be identified (Tenlen et al. 2008).Some of the phenotypes associated with loss of par-1 function are dependent on the function of mex-5 and mex-6. First, loss of function of par-1 leads to a decreased stability and aberrant localization of the posterior cell fate specification marker PIE-1, a protein that is usually inherited by the posterior daughter cell in wild-type animals and ensures the correct specification of the germline (Mello et al. 1996; Seydoux et al. 1996). This decreased stability is dependent on mex-5/6 function as PIE-1 levels are restored, albeit with symmetrical distribution, in mex-6(RNAi); mex-5(RNAi); par-1(b274) embryos (Schubert et al. 2000; Cuenca et al. 2003; Derenzo et al. 2003). Second, embryos lacking par-1 function exhibit decreased amounts of P granules in the one-cell embryo, while these markers are present in mex-6(pk440); mex-5(zu199); par-1(RNAi) embryos of comparable age (Cheeks et al. 2004). Third, in par-1(RNAi) one-cell embryos the posterior cortical domain occupied by the polarity protein PAR-2 is extended anteriorly, when compared to wild-type embryos (Cuenca et al. 2003). This anterior extension is rescued in embryos deficient for both par-1 and mex-5/6 (Cuenca et al. 2003). Taken together, these results indicate that par-1 acts in the embryo—at least in part—by regulating the localization and/or activity of the proteins MEX-5 and MEX-6. However, it remains unclear whether other proteins can modulate PAR-1 function to affect MEX-5/6 activity.To gain insight into the mechanisms of par-1 function in the embryo, we sought to identify genes that act together with par-1 during embryonic development. We performed an RNAi-based screen for genetic interactors of the temperature-sensitive allele par-1(zu310), using the embryonic lethal phenotype of this mutant as a readout. This method has proven successful in previous screens to identify genes involved in early embryonic processes (Labbé et al. 2006; O''Rourke et al. 2007). We were able to identify six genes that, upon disruption of their function, suppress the embryonic lethal phenotype of par-1 mutant embryos. One of these genes is mpk-1, the C. elegans homolog of the highly conserved MAP kinase ERK. Closer analysis subsequently showed that reduction of function of mpk-1 not only increases viability of par-1 mutant embryos, but also reverts several polarity phenotypes associated with loss of function of par-1. Our data indicate that mpk-1 antagonizes par-1 activity to regulate polarization and asymmetric cell divisions in the early embryo.  相似文献   

17.
18.
The electron transport chains in the membranes of bacteria and organelles generate proton-motive force essential for ATP production. The c-type cytochromes, defined by the covalent attachment of heme to a CXXCH motif, are key electron carriers in these energy-transducing membranes. In mitochondria, cytochromes c and c1 are assembled by the cytochrome c heme lyases (CCHL and CC1HL) and by Cyc2p, a putative redox protein. A cytochrome c1 mutant with a CAPCH heme-binding site instead of the wild-type CAACH is strictly dependent upon Cyc2p for assembly. In this context, we found that overexpression of CC1HL, as well as mutations of the proline in the CAPCH site to H, L, S, or T residues, can bypass the absence of Cyc2p. The P mutation was postulated to shift the CXXCH motif to an oxidized form, which must be reduced in a Cyc2p-dependent reaction before heme ligation. However, measurement of the redox midpoint potential of apocytochrome c1 indicates that neither the P nor the T residues impact the thermodynamic propensity of the CXXCH motif to occur in a disulfide vs. dithiol form. We show instead that the identity of the second intervening residue in the CXXCH motif is key in determining the CCHL-dependent vs. CC1HL-dependent assembly of holocytochrome c1. We also provide evidence that Cyc2p is dedicated to the CCHL pathway and is not required for the CC1HL-dependent assembly of cytochrome c1.THE c-type cytochromes, also referred to as cytochrome c, represent a universal class of heme-containing proteins that function as electron carriers in the energy-transducing pathways of bacteria, plastids, and mitochondria (Thöny-Meyer 1997; Nakamoto et al. 2000; Bonnard et al. 2010). Because cytochromes c carry a heme covalently attached to a CXXCH motif, they constitute an attractive object of study to address the question of cofactor protein assembly. The biochemical requirements for cytochrome c assembly were deduced from in vivo and in vitro studies, and the conclusion is that both apocytochromes c and heme are transported independently across at least one biological membrane and maintained as reduced prior to catalysis of the heme attachment reaction (Allen et al. 2003; Hamel et al. 2009; Kranz et al. 2009; Sanders et al. 2010). Bacterial cytochromes c are assembled in the periplasmic space, a compartment where cysteine pairs in proteins form disulfide bonds in reactions catalyzed by dedicated enzymes (Inaba 2009; Kadokura and Beckwith 2010). The current thinking holds that a c-type apocytochrome is a substrate of the disulfide bond-forming pathway, which introduces an intramolecular disulfide between the two cysteines of the CXXCH sequence (Allen et al. 2003; Sanders et al. 2010). This disulfide needs to be reduced to a dithiol to provide free sulfhydryls for the heme ligation. Consistent with this view is the fact that groups of specific oxido-reductases that constitute a transmembrane dithiol-disulfide relay from the cytosol to the periplasmic space have been shown to function as c-type cytochrome assembly factors (Allen et al. 2003; Kadokura et al. 2003; Mapller and Hederstedt 2006; Sanders et al. 2010). The proposal that the components of this pathway control the in vivo redox status of the CXXCH sulfhydryls has been inferred from the presence of motifs in their protein sequences that are consistent with a function in redox chemistry and also from the demonstration that their recombinant forms participate in dithiol–disulfide exchange reactions (Monika et al. 1997; Setterdahl et al. 2000). Moreover, the ability of exogenous thiol compounds to bypass the lack of these factors in vivo substantiates the view that the redox components have a disulfide-reducing activity in the pathway (e.g., Sambongi and Ferguson 1994; Fabianek et al. 1998; Beckett et al. 2000; Deshmukh et al. 2000; Bardischewsky and Friedrich 2001; Erlendsson and Hederstedt 2002; Erlendsson et al. 2003; Feissner et al. 2005; Turkarslan et al. 2008).While the role of these pathways is well established in bacteria, much less is known about the components that catalyze thiol/disulfide chemistry in the mitochondrial intermembrane space (IMS), which is topologically equivalent to the bacterial periplasm. By analogy with the bacterial pathways, the participation of redox-active factors that catalyze thiol formation is expected, as the mitochondrial IMS houses two c-type cytochromes, the soluble cytochrome c and the membrane-bound cytochrome c1, both of which function in respiration. In fungi, heme attachment to apocytochromes c and c1 is dependent upon the IMS resident cytochrome c and c1 heme lyases, CCHL and CC1HL, although the exact role of these lyases in the assembly process is still unclear (Dumont et al. 1987; Zollner et al. 1992). Conversion of apocytochrome to holocytochrome c depends only on CCHL, while apocytochrome c1 can be acted upon by both CCHL and CC1HL (Matner and Sherman 1982; Dumont et al. 1987; Stuart et al. 1990; Zollner et al. 1992; Bernard et al. 2003). In animals, apoforms of cytochromes c and c1 are assembled by a unique heme lyase, HCCS, which carries both the CCHL and CC1HL activities (Prakash et al. 2002; Schwarz and Cox 2002; Bernard et al. 2003).Cyc2p, a component first described as a mitochondrial biogenesis factor in yeast (Matner and Sherman 1982; Dumont et al. 1993; Pearce et al. 1998; Sanchez et al. 2001), was recently rediscovered in the context of cytochrome c1 maturation (Bernard et al. 2003). Cyc2p is located at the mitochondrial inner membrane with its C-terminal domain containing a non-covalently bound FAD exposed to the IMS (Bernard et al. 2005). A redox function for Cyc2p is likely based on the finding that a recombinant form of the molecule exhibits a NAD(P)H-dependent reductase activity (Bernard et al. 2005). However, as Cyc2p activity is not essential for the maturation process, a functional redundancy was postulated based on the fact that a cyc2-null mutant still assembles holoforms of cytochromes c and c1 (Bernard et al. 2005). The absolute requirement of Cyc2p was revealed via genetic analysis of the cyc2-null cyt1-34 combination that displays a synthetic respiratory-deficient phenotype with loss of holocytochrome c1 assembly (Bernard et al. 2005). The cyt1-34 mutation maps to the gene encoding cytochrome c1 and results in a CAPCH heme-binding site replacing the wild-type CAACH site (Bernard et al. 2005). The synthetic interaction is specific for the cyt1-34 allele carrying the A-to-P mutation and is not observed in a cyc2-null cyt1-48 strain carrying an A-to-D mutation at the heme-binding site of apocytochrome c1 (Bernard et al. 2005). The fact that Cyc2p becomes essential when the cytochrome c1 heme-binding site carries an A-to-P mutation suggests that the CXXCH motif could be the target of Cyc2p action in vivo. One possible interpretation for this observation is that the P residue alters the reactivity of the cysteinyl thiols to redox chemistry so that the apocytochrome c1 CAPCH heme-binding site occurs in an oxidized (disulfide) form, which must be reduced in a Cyc2p-dependent reaction before heme attachment can occur.In this article, we have undertaken a genetic approach to elucidate this pathway and searched for suppressors that alleviate the respiratory deficiency of the cyc2-null cyt1-34 strain. Either overexpression of CC1HL or replacement of the P mutation in the heme-binding site by H, L, S, or T residues restore the assembly of holocytochrome c1. In vitro measurement of redox potential of apoforms of CA(A/P/T)CH cytochrome c1 indicates that there is no change in the thermodynamic stability of the disulfide at the CXXCH motif that could account for the Cyc2p-dependent assembly of cytochrome c1. Genetic studies reveal that the replacement of the second A residue at the CAACH motif by H, L, P, S, and T residues is key in determining the conversion of apocytochrome c1 to its corresponding holoform via the CCHL and/or CC1HL-dependent pathway. We also demonstrate that Cyc2p is a component dedicated to the CCHL pathway and is not required for the CC1HL-dependent assembly of cytochrome c1. We propose that the CAPCH cytochrome c1 is strictly dependent upon CCHL and Cyc2p for its assembly but becomes a substrate of CC1HL upon overexpression of CC1HL or in the presence of H, L, S, or T mutations.  相似文献   

19.
20.
Gross chromosomal rearrangements (GCRs) are large scale changes to chromosome structure and can lead to human disease. We previously showed in Saccharomyces cerevisiae that nearby inverted repeat sequences (∼20–200 bp of homology, separated by ∼1–5 kb) frequently fuse to form unstable dicentric and acentric chromosomes. Here we analyzed inverted repeat fusion in mutants of three sets of genes. First, we show that genes in the error-free postreplication repair (PRR) pathway prevent fusion of inverted repeats, while genes in the translesion branch have no detectable role. Second, we found that siz1 mutants, which are defective for Srs2 recruitment to replication forks, and srs2 mutants had opposite effects on instability. This may reflect separate roles for Srs2 in different phases of the cell cycle. Third, we provide evidence for a faulty template switch model by studying mutants of DNA polymerases; defects in DNA pol delta (lagging strand polymerase) and Mgs1 (a pol delta interacting protein) lead to a defect in fusion events as well as allelic recombination. Pol delta and Mgs1 may collaborate either in strand annealing and/or DNA replication involved in fusion and allelic recombination events. Fourth, by studying genes implicated in suppression of GCRs in other studies, we found that inverted repeat fusion has a profile of genetic regulation distinct from these other major forms of GCR formation.ALL organisms are prone to large-scale changes (gross chromosomal rearrangements, GCRs) to their genomes that include deletions, inversions, and translocations. These large-scale changes are thought to drive evolutionary events, such as speciation, and contribute to human pathology such as Pelziaeus-Merzbacher syndrome and other genetic disorders (Lee et al. 2007; Stankiewicz and Lupski 2010). Thus, a firm understanding of how cells normally prevent such rearrangements, and how they accumulate, is critical to our understanding of both evolution and pathology.GCRs arise by many different mechanisms, and there is growing evidence that errors during DNA replication are a major source (Myung et al. 2001; Admire et al. 2006; Mizuno et al. 2009). Errors are thought to arise when replication forks encounter “lesions” on the template strand. Lesions can consist of protein complexes bound to DNA or lesions in the DNA itself. Replication forks bypass lesions by several different mechanisms that are still poorly understood (Atkinson and McGlynn 2009; Weinert et al. 2009). We believe that understanding lesion bypass mechanisms is central to understanding both how GCRs are prevented and how they form when lesion bypass mechanisms fail.All lesion bypass pathways utilize sequence homology to restart replication (Atkinson and McGlynn 2009; Weinert et al. 2009). Use of sequence homology during restart may limit the frequency of GCRs, as it lowers the probability of annealing to nonallelic sequences. Repetitive sequences present a problem because lesion bypass at sites near repetitive sequences may lead to annealing of nonallelic sequences and thus to GCR formation (Lemoine et al. 2005; Narayanan et al. 2006; Argueso et al. 2008). Indeed in yeast and in other organisms, GCRs occur frequently in repeat sequences (Dunham et al. 2002; Argueso et al. 2008; Di Rienzi et al. 2009). Some rearrangements do occur between so-called “single-copy sequences” with either no homology or limited homology (microhomologies of 5–9 bp; Myung et al. 2001; Kolodner et al. 2002; Putnam et al. 2005) though evidence suggests these rearrangements occur less frequently than rearrangements between repetitive sequences (Putnam et al. 2009). Interestingly, it has been shown that some genes are required to prevent the fusion of repetitive elements yet have no effect on rearrangements between single-copy sequences (Putnam et al. 2009). Currently it is not clear how these pathways act to suppress repeat-mediated events and why they are not required to prevent rearrangements between single-copy sequences.Our current understanding of the mechanisms underlying GCR formation is mostly derived from assays designed to detect specific changes to yeast chromosomes (Chen and Kolodner 1999; Myung et al. 2001; Huang and Koshland 2003; Lambert et al. 2005; Rattray et al. 2005; Admire et al. 2006; Narayanan et al. 2006; Schmidt et al. 2006; Smith et al. 2007; Pannunzio et al. 2008; Payen et al. 2008; Paek et al. 2009; Mizuno et al. 2009). Previously we reported on GCR formation in the budding yeast Saccharomyces cerevisiae using an assay we developed. We found that a major source of genome instability involves the fusion of nearby inverted repeats (with ∼20–200 bp of sequence homology, separated by 1–5 kb) to form either dicentric or acentric chromosomes (Figure 1D; Paek et al. 2009). We also found that fusion of inverted repeats is general: fusion occurred between inverted repeats at all five different locations tested on four different yeast chromosomes, as well as between synthetic inverted repeats (Paek et al. 2009). Genetic data suggest that these events most likely occur during replication of DNA (Admire et al. 2006). Further genetic analysis suggested that the mechanism of inverted repeat fusion differed from that of direct repeat recombination, in that inverted repeat fusion did not require genes involved in homologous recombination (HR) or single-strand annealing (SSA) pathways (Paek et al. 2009). In addition, fusion events are unlikely to involve double-strand breaks (DSBs), as genes in the nonhomologous end joining (NHEJ) and microhomology-mediated end joining (MMEJ) are not required for fusion events (Paek et al. 2009). Indeed gene knockouts in the HR (RAD52, RAD51, and RAD59), SSA (RAD52 and RAD1) and postreplication repair (PRR) (RAD18) pathways actually increased the frequency of fusion of an inverted repeat on chromosome (Chr) VII (Paek et al. 2009); these pathways normally suppress inverted repeat fusion.Open in a separate windowFigure 1.—Experimental setup for the detection of inverted repeat fusion and chromosome instability. Objects are not drawn to scale. (A) The starting strain has two copies of Chr VII. One copy contains the CAN1 gene, ADE6, ade3, while the other copy is ade6, ADE3. Cells are plated to canavanine, and three types of colonies are formed: (B) Allelic recombinants are round in appearance and are Ade+; (C) colonies that form by loss of Chr VII are round in appearance and Ade; and (D) cells that contain unstable dicentric chromosomes form by the fusion of inverted repeats. One specific case of this fusion (the S2/S3 dicentric) is shown within braces. Cells with dicentrics form mixed colonies, which contain allelic recombinants, chromosome loss events, as well as a translocation between D7 and D11. The bar in the S2/S3 repeat represents a fusion junction. (E) The specific dicentric is detected by dicentric primers DP1 and DP2 and (F) a monocentric translocation that is detected with translocation primers TP1 and TP2.To further our previous studies, we analyzed three groups of genes implicated in the maintenance of genome stability. We tested how these genes affect the overall stability of Chr VII, focusing on the fusion of nearby inverted repeats to form a specific dicentric Chr VII and the resolution of the dicentric into a monocentric translocation (which we term the 403–535 translocation; Figure 1, D–F). First, we analyzed several genes in the PRR pathway and found that error-free bypass, but not translesion synthesis, is required for the prevention of inverted repeat fusion. Surprisingly, we found that siz1 mutants, which are defective for Srs2 recruitment to replication forks, and srs2 mutants had opposite effects on instability. This may reflect separate roles for Srs2 in different phases of the cell cycle. Second, we analyzed several mutations in genes that are associated with replication forks. We found that mutants in POL3 (polymerase delta) and MGS1 (encoding a single-strand annealing protein, which binds polymerase delta) significantly reduced the frequency of dicentric formation and allelic recombinants that arise in the checkpoint mutant rad9 (Giot et al. 1997; Hishida et al. 2001; Paek et al. 2009). Finally we studied genes associated with rearrangements involving repeats or single-copy sequences, as well as a subset of mutants involved in recombination. Generally, we find that the mechanisms of nearby inverted repeat fusion are distinct from mechanisms fusing longer repeats or single-copy sequences.  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