27 research outputs found

    A common, non-optimal phenotypic endpoint in experimental adaptations of bacteriophage lysis time

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Optimality models of evolution, which ignore genetic details and focus on natural selection, are widely used but sometimes criticized as oversimplifications. Their utility for quantitatively predicting phenotypic evolution can be tested experimentally. One such model predicts optimal bacteriophage lysis interval, how long a virus should produce progeny before lysing its host bacterium to release them. The genetic basis of this life history trait is well studied in many easily propagated phages, making it possible to test the model across a variety of environments and taxa.</p> <p>Results</p> <p>We adapted two related small single-stranded DNA phages, ΦX174 and ST-1, to various conditions. The model predicted the evolution of the lysis interval in response to host density and other environmental factors. In all cases the initial phages lysed later than predicted. The ΦX174 lysis interval did not evolve detectably when the phage was adapted to normal hosts, indicating complete failure of optimality predictions. ΦX174 grown on slyD-defective hosts which initially entirely prevented lysis readily recovered to a lysis interval similar to that attained on normal hosts. Finally, the lysis interval still evolved to the same endpoint when the environment was altered to delay optimal lysis interval. ST-1 lysis interval evolved to be ~2 min shorter, qualitatively in accord with predictions. However, there were no changes in the single known lysis gene. Part of ST-1's total lysis time evolution consisted of an earlier start to progeny production, an unpredicted phenotypic response outside the boundaries of the optimality model.</p> <p>Conclusions</p> <p>The consistent failure of the optimality model suggests that constraint and genetic details affect quantitative and even qualitative success of optimality predictions. Several features of ST-1 adaptation show that lysis time is best understood as an output of multiple traits, rather than in isolation.</p

    The Second-Shell Metal Ligands of Human Arginase Affect Coordination of the Nucleophile and Substrate†

    Get PDF
    ABSTRACT: The active sites of eukaryotic arginase enzymes are strictly conserved, especially the first- and second-shell ligands that coordinate the two divalent metal cations that generate a hydroxide molecule for nucleophilic attack on the guanidinium carbon of L-arginine and the subsequent production of urea and L-ornithine. Here by using comprehensive pairwise saturation mutagenesis of the first- and second-shell metal ligands in human arginase I, we demonstrate that several metal binding ligands are actually quite tolerant to amino acid substitutions. Of&gt;2800 double mutants of first- and second-shell residues analyzed, we found more than 80 unique amino acid substitutions, of which four were in first-shell residues. Remarkably, certain second-shell mutations could modulate the binding of both the nucleophilic water/hydroxide molecule and substrate or product ligands, resulting in activity greater than that of the wild-type enzyme. The data presented here constitute the first comprehensive saturation mutagenesis analysis of a metallohydrolase active site and reveal that the strict conservation of the second-shell metal binding residues in eukaryotic arginases does not reflect kinetic optimization of the enzyme during the course of evolution. Arginases (EC 3.5.3.1) are typically homotrimeric enzymes with an R/β fold comprising an eight-strand β-sheet surrounded by several helices. The enzyme contains a dinuclear metal center tha

    Mechanism of arginine sensing by CASTOR1 upstream of mTORC1

    Get PDF
    The mechanistic Target of Rapamycin Complex 1 (mTORC1) is a major regulator of eukaryotic growth that coordinates anabolic and catabolic cellular processes with inputs such as growth factors and nutrients, including amino acids. In mammals arginine is particularly important, promoting diverse physiological effects such as immune cell activation, insulin secretion, and muscle growth, largely mediated through activation of mTORC1 (refs 4, 5, 6, 7).Arginine activates mTORC1 upstream of the Rag family of GTPases, through either the lysosomal amino acid transporter SLC38A9 or the GATOR2-interacting Cellular Arginine Sensor for mTORC1 (CASTOR1). However, the mechanism by which the mTORC1 pathway detects and transmits this arginine signal has been elusive. Here, we present the 1.8 Ã… crystal structure of arginine-bound CASTOR1. Homodimeric CASTOR1 binds arginine at the interface of two Aspartate kinase, Chorismate mutase, TyrA (ACT) domains, enabling allosteric control of the adjacent GATOR2-binding site to trigger dissociation from GATOR2 and downstream activation of mTORC1. Our data reveal that CASTOR1 shares substantial structural homology with the lysine-binding regulatory domain of prokaryotic aspartate kinases, suggesting that the mTORC1 pathway exploited an ancient, amino-acid-dependent allosteric mechanism to acquire arginine sensitivity. Together, these results establish a structural basis for arginine sensing by the mTORC1 pathway and provide insights into the evolution of a mammalian nutrient sensor.National Institutes of Health (U.S.) (Grant R01CA103866)National Institutes of Health (U.S.) (Grant AI47389)United States. Department of Defense (Award W81XWH-07-0448)National Institutes of Health (U.S.) (Grant F31 CA180271

    Sestrin2 is a leucine sensor for the mTORC1 pathway

    Get PDF
    Leucine is a proteogenic amino acid that also regulates many aspects of mammalian physiology, in large part by activating the mTOR complex 1 (mTORC1) protein kinase, a master growth controller. Amino acids signal to mTORC1 through the Rag guanosine triphosphatases (GTPases). Several factors regulate the Rags, including GATOR1, aGTPase-activating protein; GATOR2, a positive regulator of unknown function; and Sestrin2, a GATOR2-interacting protein that inhibits mTORC1 signaling. We find that leucine, but not arginine, disrupts the Sestrin2-GATOR2 interaction by binding to Sestrin2 with a dissociation constant of 20 micromolar, which is the leucine concentration that half-maximally activates mTORC1. The leucine-binding capacity of Sestrin2 is required for leucine to activate mTORC1 in cells. These results indicate that Sestrin2 is a leucine sensor for the mTORC1 pathway.United States. National Institutes of Health (R01CA103866)United States. National Institutes of Health (AI47389)United States. Department of Defense (W81XWH-07-0448)United States. National Institutes of Health (T32 GM007753)United States. National Institutes of Health (F30 CA189333)United States. National Institutes of Health (F31 CA180271

    The Folliculin Tumor Suppressor Is a GAP for the RagC/D GTPases That Signal Amino Acid Levels to mTORC1

    Get PDF
    The mTORC1 kinase is a master growth regulator that senses numerous environmental cues, including amino acids. The Rag GTPases interact with mTORC1 and signal amino acid sufficiency by promoting the translocation of mTORC1 to the lysosomal surface, its site of activation. The Rags are unusual GTPases in that they function as obligate heterodimers, which consist of RagA or B bound to RagC or D. While the loading of RagA/B with GTP initiates amino acid signaling to mTORC1, the role of RagC/D is unknown. Here, we show that RagC/D is a key regulator of the interaction of mTORC1 with the Rag heterodimer and that, unexpectedly, RagC/D must be GDP bound for the interaction to occur. We identify FLCN and its binding partners, FNIP1/2, as Rag-interacting proteins with GAP activity for RagC/D, but not RagA/B. Thus, we reveal a role for RagC/D in mTORC1 activation and a molecular function for the FLCN tumor suppressor.United States. National Institutes of Health (CA103866)United States. National Institutes of Health (AI47389)United States. Department of Defense (W81XWH-07-0448)National Cancer Institute (U.S.) (F30CA180754

    Structural basis for leucine sensing by the Sestrin2-mTORC1 pathway

    Get PDF
    Eukaryotic cells coordinate growth with the availability of nutrients through the mechanistic target of rapamycin complex 1 (mTORC1), a master growth regulator. Leucine is of particular importance and activates mTORC1 via the Rag guanosine triphosphatases and their regulators GATOR1 and GATOR2. Sestrin2 interacts with GATOR2 and is a leucine sensor. Here we present the 2.7 angstrom crystal structure of Sestrin2 in complex with leucine. Leucine binds through a single pocket that coordinates its charged functional groups and confers specificity for the hydrophobic side chain. A loop encloses leucine and forms a lid-latch mechanism required for binding. A structure-guided mutation in Sestrin2 that decreases its affinity for leucine leads to a concomitant increase in the leucine concentration required for mTORC1 activation in cells. These results provide a structural mechanism of amino acid sensing by the mTORC1 pathway.United States. Department of Defense (W81XWH-07- 0448)Damon Runyon Cancer Research Foundation (DRG-112-12)National Institutes of Health (U.S.) (Predoctoral Training Grant T32GM007287)National Institutes of Health (U.S.) (Grants R01CA103866, AI47389, T32 GM007753, F30 CA189333, F31 CA180271, and F31 CA189437)United States. Dept. of Defense. Breast Cancer Research Program (Postdoctoral Fellowship BC120208)Massachusetts Institute of Technology. Office of the Dean for Graduate Education (Whitaker Health Sciences Fund Fellowship)Damon Runyon Cancer Research Foundation (Sally Gordon Fellowship DRG-112-12

    The CASTOR Proteins Are Arginine Sensors for the mTORC1 Pathway

    Get PDF
    Amino acids signal to the mTOR complex I (mTORC1) growth pathway through the Rag GTPases. Multiple distinct complexes regulate the Rags, including GATOR1, a GTPase activating protein (GAP), and GATOR2, a positive regulator of unknown molecular function. Arginine stimulation of cells activates mTORC1, but how it is sensed is not well understood. Recently, SLC38A9 was identified as a putative lysosomal arginine sensor required for arginine to activate mTORC1 but how arginine deprivation represses mTORC1 is unknown. Here, we show that CASTOR1, a previously uncharacterized protein, interacts with GATOR2 and is required for arginine deprivation to inhibit mTORC1. CASTOR1 homodimerizes and can also heterodimerize with the related protein, CASTOR2. Arginine disrupts the CASTOR1-GATOR2 complex by binding to CASTOR1 with a dissociation constant of ∼30 μM, and its arginine-binding capacity is required for arginine to activate mTORC1 in cells. Collectively, these results establish CASTOR1 as an arginine sensor for the mTORC1 pathway.United States. National Institutes of Health (R01CA103866)United States. National Institutes of Health (AI47389)United States. Department of Energy (W81XWH-07-0448)United States. National Institutes of Health (F31 CA180271)United States. National Institutes of Health (F31 CA189437

    SCHEMA-Designed Variants of Human Arginase I and II Reveal Sequence Elements Important to Stability and Catalysis

    Get PDF
    Arginases catalyze the divalent cation-dependent hydrolysis of l-arginine to urea and l-ornithine. There is significant interest in using arginase as a therapeutic anti-neogenic agent against l-arginine auxotrophic tumors and in enzyme replacement therapy for treating hyperargininemia. Both therapeutic applications require enzymes with sufficient stability under physiological conditions. To explore sequence elements that contribute to arginase stability we used SCHEMA-guided recombination to design a library of chimeric enzymes composed of sequence fragments from the two human isozymes Arginase I and II. We then developed a novel active learning algorithm that selects sequences from this library that are both highly informative and functional. Using high-throughput gene synthesis and our two-step active learning algorithm, we were able to rapidly create a small but highly informative set of seven enzymatically active chimeras that had an average variant distance of 40 mutations from the closest parent arginase. Within this set of sequences, linear regression was used to identify the sequence elements that contribute to the long-term stability of human arginase under physiological conditions. This approach revealed a striking correlation between the isoelectric point and the long-term stability of the enzyme to deactivation under physiological conditions

    KICSTOR recruits GATOR1 to the lysosome and is necessary for nutrients to regulate mTORC1

    Get PDF
    The mechanistic target of rapamycin complex 1 kinase (mTORC1) is a central regulator of cell growth that responds to diverse environmental signals and is deregulated in many human diseases, including cancer and epilepsy1–3. Amino acids are a key input, and act through the Rag GTPases to promote the translocation of mTORC1 to the lysosomal surface, its site of activation4. Multiple protein complexes regulate the Rag GTPases in response to amino acids, including GATOR1, a GTPase activating protein for RagA, and GATOR2, a positive regulator of unknown molecular function. Here, we identify a four-membered protein complex (KICSTOR) composed of the KPTN, ITFG2, C12orf66, and SZT2 gene products as required for amino acid or glucose deprivation to inhibit mTORC1 in cultured cells. In mice lacking SZT2, mTORC1 signaling is increased in several tissues, including in neurons in the brain. KICSTOR localizes to lysosomes; binds to GATOR1 and recruits it, but not GATOR2, to the lysosomal surface; and is necessary for the interaction of GATOR1 with its substrates, the Rag GTPases, and with GATOR2. Interestingly, several KICSTOR components are mutated in neurological diseases associated with mutations that lead to hyperactive mTORC1 signaling5–10. Thus, KICSTOR is a lysosome-associated negative regulator of mTORC1 signaling that, like GATOR1, is mutated in human disease11,12

    Discovering regulators of the amino acid sensing pathway of mTORC1

    No full text
    Thesis: Ph. D., Massachusetts Institute of Technology, Department of Biology, February 2017.Cataloged from PDF version of thesis. "February 2017."Includes bibliographical references.The mechanistic target of rapamycin complex I (mTORC1) protein kinase functions as a master regulator of growth, and its deregulation is common in human disease, including cancer and diabetes. mTORC1 integrates multiple environmental cues to control anabolic and catabolic processes. A key input is amino acids, which function to promote the translocation of mTORC1 to the lysosomal surface, its site of activation. Necessary for this recruitment are the Rag GTPases and several distinct factors that modulate their nucleotide state in response to amino acid availability. Despite these advances, several key questions remain. The components that mediate mTORC1 inhibition upon amino acid deprivation and the identities of the amino acid sensors upstream of mTORC1 are both unknown. To provide insight into these questions, we undertook an unbiased proteomics approach to discover novel mTORC1 regulators. Here, we describe the identification of GATOR2 as a pentameric complex that positively regulates mTORC1 and functions upstream of or in parallel to GATOR1, a GTPase activating protein complex for the Rags and a negative regulator of the mTORC1 pathway. KICSTOR, a four-membered protein complex, is necessary to localize GATOR1 to the lysosome to enable it to suppress mTORC1 activity. GATOR1 components are mutated in cancer and may identify tumors that respond to clinically approved mTORC1 inhibitors. Furthermore, we describe the identification of Sestrin2 and CASTOR1 as GATOR2-interacting proteins that function as leucine and arginine sensors, respectively, for the mTORCI pathway. Both sensors are required to signal the absence of leucine and arginine to mTORC1, and the amino acid-binding capacity of both sensors is necessary for amino acids to activate mTORC1. Altogether, the identification of these mTORC1 regulators furthers our understanding of the mechanisms by which amino acid availability controls cellular growth.by Lynne Chantranupong.Ph. D
    corecore