Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Mol Biol Cell ; 33(13)2022 11 01.
Article in English | MEDLINE | ID: mdl-36228182

ABSTRACT

METEI (Medical Expedition to Easter Island) was a Canadian-led expedition to Easter Island in 1964 that led to the discovery of rapamycin, launching a billion-dollar drug industry and major field of biomedical research. Stanley's Dream, by medical historian Jacalyn Duffin, provides remarkable details about METEI and raises important and timely questions about systemic bias in biomedical studies, the relationship between science and geopolitics, as well as obligations of pharmaceutical companies to indigenous communities. As such, this book is a must-read for those interested in the intersection of science and society as well as anyone who has used rapamycin, or one of many derivatives, in their laboratory or clinic.


Subject(s)
Biomedical Research , Sirolimus , Canada , Pharmaceutical Preparations , Politics , Sirolimus/pharmacology
2.
Mol Biol Cell ; 33(5): ar44, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35293776

ABSTRACT

The mammalian target of rapamycin (mTOR) is a large protein kinase that assembles into two multisubunit protein complexes, mTORC1 and mTORC2, to regulate cell growth in eukaryotic cells. While significant progress has been made in our understanding of the composition and structure of these complexes, important questions remain regarding the role of specific sequences within mTOR important for complex formation and activity. To address these issues, we have used a molecular genetic approach to explore TOR complex assembly in budding yeast, where two closely related TOR paralogues, TOR1 and TOR2, partition preferentially into TORC1 versus TORC2, respectively. We previously identified an ∼500-amino-acid segment within the N-terminal half of each protein, termed the major assembly specificity (MAS) domain, which can govern specificity in formation of each complex. In this study, we have extended the use of chimeric TOR1-TOR2 genes as a "sensitized" genetic system to identify specific subdomains rendered essential for TORC2 function, using synthetic lethal interaction analyses. Our findings reveal important design principles underlying the dimeric assembly of TORC2 as well as identifying specific segments within the MAS domain critical for TORC2 function, to a level approaching single-amino-acid resolution. Together these findings highlight the complex and cooperative nature of TOR complex assembly and function.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Cell Cycle Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
3.
Science ; 360(6395)2018 06 22.
Article in English | MEDLINE | ID: mdl-29930109

ABSTRACT

Shlezinger et al (Reports, 8 September 2017, p. 1037) report that the common fungus Aspergillus fumigatus, a cause of aspergillosis, undergoes caspase-dependent apoptosis-like cell death triggered by lung neutrophils. However, the technologies they used do not provide reliable evidence that fungal cells die via a protease signaling cascade thwarted by a fungal caspase inhibitor homologous to human survivin.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/immunology , Apoptosis/immunology , Cell Death , Humans , Lung/immunology
4.
Biomolecules ; 8(2)2018 06 01.
Article in English | MEDLINE | ID: mdl-29865216

ABSTRACT

TOR is a serine/threonine protein kinase that assembles into distinct TOR Complexes 1 and 2 (TORC1 or TORC2) to regulate cell growth. In mammalian cells, a single mTOR incorporates stably into mTORC1 and mTORC2. By contrast, in Saccharomyces cerevisiae, two highly similar Tor1 and Tor2 proteins exist, where Tor1 assembles exclusively into TORC1 and Tor2 assembles preferentially into TORC2. To gain insight into TOR complex assembly, we used this bifurcation in yeast to identify structural elements within Tor1 and Tor2 that govern their complex specificity. We have identified a concise region of ~500 amino acids within the N-terminus of Tor2, which we term the Major Assembly Specificity (MAS) domain, that is sufficient to confer significant TORC2 activity when placed into an otherwise Tor1 protein. Consistently, introduction of the corresponding MAS domain from Tor1 into an otherwise Tor2 is sufficient to confer stable association with TORC1-specific components. Remarkably, much like mTOR, this latter chimera also retains stable interactions with TORC2 components, indicating that determinants throughout Tor1/Tor2 contribute to complex specificity. Our findings are in excellent agreement with recent ultrastructural studies of TORC1 and TORC2, where the MAS domain is involved in quaternary interactions important for complex formation and/or stability.


Subject(s)
Cell Cycle Proteins/chemistry , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Phosphatidylinositol 3-Kinases/chemistry , Protein Multimerization , Saccharomyces cerevisiae Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Protein Domains , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
5.
Autophagy ; 13(11): 1804-1812, 2017.
Article in English | MEDLINE | ID: mdl-29198169

ABSTRACT

Macroautophagy/autophagy is a starvation and stress-induced catabolic process critical for cellular homeostasis and adaptation. Several Atg proteins are involved in the formation of the autophagosome and subsequent degradation of cytoplasmic components, a process termed autophagy flux. Additionally, the expression of several Atg proteins, in particular Atg8, is modulated transcriptionally, yet the regulatory mechanisms involved remain poorly understood. Here we demonstrate that the AGC kinase Ypk1, target of the rapamycin-insensitive TORC2 signaling pathway, controls ATG8 expression by repressing the heterodimeric Zinc-finger transcription factors Msn2 and Msn4. We find that Msn2 and Msn4 promote ATG8 expression downstream of the histone deacetylase complex (HDAC) subunit Ume6, a previously identified negative regulator of ATG8 expression. Moreover, we demonstrate that TORC2-Ypk1 signaling is functionally linked to distinct mitochondrial respiratory complexes. Surprisingly, we find that autophagy flux during amino acid starvation is also dependent upon Msn2-Msn4 activity, revealing a broad role for these transcription factors in the autophagy response.


Subject(s)
Autophagy-Related Protein 8 Family/genetics , Autophagy/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Fungal , Glycogen Synthase Kinase 3/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Amino Acids/deficiency , Mitochondria/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae/genetics , Signal Transduction , Stress, Physiological/genetics
6.
J Cell Biol ; 216(9): 2679-2689, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28774891

ABSTRACT

Membrane contact sites (MCSs) function to facilitate the formation of membrane domains composed of specialized lipids, proteins, and nucleic acids. In cells, membrane domains regulate membrane dynamics and biochemical and signaling pathways. We and others identified a highly conserved family of sterol transport proteins (Ltc/Lam) localized at diverse MCSs. In this study, we describe data indicating that the yeast family members Ltc1 and Ltc3/4 function at the vacuole and plasma membrane, respectively, to create membrane domains that partition upstream regulators of the TORC1 and TORC2 signaling pathways to coordinate cellular stress responses with sterol homeostasis.


Subject(s)
Antiporters/metabolism , Membrane Microdomains/enzymology , Multiprotein Complexes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Sterols/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Antiporters/genetics , Biological Transport , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cytoskeletal Proteins , Endoplasmic Reticulum/enzymology , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Vacuoles/enzymology
7.
Autophagy ; 13(7): 1256-1257, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28324658

ABSTRACT

The target of rapamycin (TOR) kinase is a conserved regulator of cell growth and functions within 2 different protein complexes, TORC1 and TORC2, where TORC2 positively controls macroautophagy/autophagy during amino acid starvation. Under these conditions, TORC2 signaling inhibits the activity of the calcium-regulated phosphatase calcineurin and promotes the general amino acid control (GAAC) response and autophagy. Here we demonstrate that TORC2 regulates calcineurin by controlling the respiratory activity of mitochondria. In particular, we find that mitochondrial oxidative stress affects the calcium channel regulatory protein Mid1, which we show is an essential upstream activator of calcineurin. Thus, these findings describe a novel regulation for autophagy that involves TORC2 signaling, mitochondrial respiration, and calcium homeostasis.


Subject(s)
Autophagy , Calcium/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mitochondria/metabolism , Calcineurin/metabolism , Cell Respiration , Glycogen Synthase Kinase 3/metabolism , Membrane Glycoproteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction
8.
Curr Genet ; 63(1): 35-42, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27233284

ABSTRACT

In eukaryotic cells, cellular homeostasis requires that different organelles respond to intracellular as well as environmental signals and modulate their behavior as conditions demand. Understanding the molecular mechanisms required for these changes remains an outstanding goal. One such organelle is the lysosome/vacuole, which undergoes alterations in size and number in response to environmental and physiological stimuli. Changes in the morphology of this organelle are mediated in part by the equilibrium between fusion and fission processes. While the fusion of the yeast vacuole has been studied intensively, the regulation of vacuolar fission remains poorly characterized by comparison. In recent years, a number of studies have incorporated genome-wide visual screens and high-throughput microscopy to identify factors required for vacuolar fission in response to diverse cellular insults, including hyperosmotic and endoplasmic reticulum stress. Available evidence now demonstrates that the rapamycin-sensitive TOR network, a master regulator of cell growth, is required for vacuolar fragmentation in response to stress. Importantly, many of the genes identified in these studies provide new insights into potential links between the vacuolar fission machinery and TOR signaling. Together these advances both extend our understanding of the regulation of vacuolar fragmentation in yeast as well as underscore the role of analogous events in mammalian cells.


Subject(s)
Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Vacuoles/metabolism , Animals , Biological Transport , Gene Expression Regulation , Humans , Intracellular Membranes/metabolism , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Protein Binding , Protein Transport , Stress, Physiological , Yeasts/genetics , Yeasts/metabolism
9.
J Cell Biol ; 215(6): 779-788, 2016 Dec 19.
Article in English | MEDLINE | ID: mdl-27899413

ABSTRACT

Autophagy is a catabolic process that recycles cytoplasmic contents and is crucial for cell survival during stress. The target of rapamycin (TOR) kinase regulates autophagy as part of two distinct protein complexes, TORC1 and TORC2. TORC1 negatively regulates autophagy according to nitrogen availability. In contrast, TORC2 functions as a positive regulator of autophagy during amino acid starvation, via its target kinase Ypk1, by repressing the activity of the calcium-dependent phosphatase calcineurin and promoting the general amino acid control (GAAC) response. Precisely how TORC2-Ypk1 signaling regulates calcineurin within this pathway remains unknown. Here we demonstrate that activation of calcineurin requires Mid1, an endoplasmic reticulum-localized calcium channel regulatory protein implicated in the oxidative stress response. We find that normal mitochondrial respiration is perturbed in TORC2-Ypk1-deficient cells, which results in the accumulation of mitochondrial-derived reactive oxygen species that signal to Mid1 to activate calcineurin, thereby inhibiting the GAAC response and autophagy. These findings describe a novel pathway involving TORC2, mitochondrial oxidative stress, and calcium homeostasis for autophagy regulation.


Subject(s)
Autophagy , Calcium Channels/metabolism , Membrane Glycoproteins/metabolism , Mitochondria/metabolism , Multiprotein Complexes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , TOR Serine-Threonine Kinases/metabolism , Amino Acids/metabolism , Calcineurin/metabolism , Cell Respiration , Mechanistic Target of Rapamycin Complex 2 , Models, Biological
10.
Mol Biol Cell ; 26(25): 4618-30, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26466677

ABSTRACT

The yeast vacuole is equivalent to the mammalian lysosome and, in response to diverse physiological and environmental stimuli, undergoes alterations both in size and number. Here we demonstrate that vacuoles fragment in response to stress within the endoplasmic reticulum (ER) caused by chemical or genetic perturbations. We establish that this response does not involve known signaling pathways linked previously to ER stress but instead requires the rapamycin-sensitive TOR Complex 1 (TORC1), a master regulator of cell growth, together with its downstream effectors, Tap42/Sit4 and Sch9. To identify additional factors required for ER stress-induced vacuolar fragmentation, we conducted a high-throughput, genome-wide visual screen for yeast mutants that are refractory to ER stress-induced changes in vacuolar morphology. We identified several genes shown previously to be required for vacuolar fusion and/or fission, validating the utility of this approach. We also identified a number of new components important for fragmentation, including a set of proteins involved in assembly of the V-ATPase. Remarkably, we find that one of these, Vph2, undergoes a change in intracellular localization in response to ER stress and, moreover, in a manner that requires TORC1 activity. Together these results reveal a new role for TORC1 in the regulation of vacuolar behavior.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Transcription Factors/genetics , Vacuoles/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Proliferation/genetics , Endoplasmic Reticulum/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Vacuoles/metabolism
11.
J Biol Chem ; 290(3): 1395-403, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25429105

ABSTRACT

Complex sphingolipids are important components of eukaryotic cell membranes and, together with their biosynthetic precursors, including sphingoid long chain bases and ceramides, have important signaling functions crucial for cell growth and survival. Ceramides are produced at the endoplasmic reticulum (ER) membrane by a multicomponent enzyme complex termed ceramide synthase (CerS). In budding yeast, this complex is composed of two catalytic subunits, Lac1 and Lag1, as well as an essential regulatory subunit, Lip1. Proper formation of ceramides by CerS has been shown previously to require the Cka2 subunit of casein kinase 2 (CK2), a ubiquitous enzyme with multiple cellular functions, but the precise mechanism involved has remained unidentified. Here we present evidence that Lac1 and Lag1 are direct targets for CK2 and that phosphorylation at conserved positions within the C-terminal cytoplasmic domain of each protein is required for optimal CerS activity. Our data suggest that phosphorylation of Lac1 and Lag1 is important for proper localization and distribution of CerS within the ER membrane and that phosphorylation of these sites is functionally linked to the COP I-dependent C-terminal dilysine ER retrieval pathway. Together, our data identify CK2 as an important regulator of sphingolipid metabolism, and additionally, because both ceramides and CK2 have been implicated in the regulation of cancer, our findings may lead to an enhanced understanding of their relationship in health and disease.


Subject(s)
Casein Kinase II/metabolism , Gene Expression Regulation, Fungal , Membrane Proteins/metabolism , Oxidoreductases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Amino Acid Motifs , Amino Acid Sequence , Catalytic Domain , Ceramides/metabolism , Endoplasmic Reticulum/metabolism , Humans , Microscopy, Fluorescence , Molecular Sequence Data , Phosphorylation , Plasmids/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Sphingolipids/chemistry
12.
Autophagy ; 10(11): 2085-6, 2014.
Article in English | MEDLINE | ID: mdl-25426890

ABSTRACT

The conserved target of rapamycin (TOR) kinase is a central regulator of cell growth in response to nutrient availability. TOR forms 2 structurally and functionally distinct complexes, TORC1 and TORC2, and negatively regulates autophagy via TORC1. Here we demonstrate TOR also operates independently through the TORC2 signaling pathway to promote autophagy upon amino acid limitation. Under these conditions, TORC2, through its downstream target kinase Ypk1, inhibits the Ca(2+)- and Cmd1/calmodulin-dependent phosphatase, calcineurin, to enable the activation of the amino acid-sensing EIF2S1/eIF2α kinase, Gcn2, and promote autophagy. Thus TORC2 signaling regulates autophagy in a pathway distinct from TORC1 to provide a tunable response to the cellular metabolic state.


Subject(s)
Autophagy , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Amino Acids/chemistry , Calcineurin/metabolism , Calcium/chemistry , Calcium/metabolism , Disease Progression , Glycogen Synthase Kinase 3/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Nitrogen/chemistry , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction
13.
Mol Biol Cell ; 25(24): 3962-72, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25253719

ABSTRACT

The evolutionarily conserved mTOR complex 2 (mTORC2) signaling pathway is an important regulator of actin cytoskeletal architecture and, as such, is a candidate target for preventing cancer cell motility and invasion. Remarkably, the precise mechanism(s) by which mTORC2 regulates the actin cytoskeleton have remained elusive. Here we show that in budding yeast, TORC2 and its downstream kinase Ypk1 regulate actin polarization by controlling reactive oxygen species (ROS) accumulation. Specifically, we find that TORC2-Ypk1 regulates actin polarization both by vacuole-related ROS, controlled by the phospholipid flippase kinase Fpk1 and sphingolipids, and by mitochondria-mediated ROS, controlled by the PKA subunit Tpk3. In addition, we find that the protein kinase C (Pkc1)/MAPK cascade, a well-established regulator of actin, acts downstream of Ypk1 to regulate ROS, in part by promoting degradation of the oxidative stress responsive repressor, cyclin C. Furthermore, we show that Ypk1 regulates Pkc1 activity through proper localization of Rom2 at the plasma membrane, which is also dependent on Fpk1 and sphingolipids. Together these findings demonstrate important links between TORC2/Ypk1 signaling, Fpk1, sphingolipids, Pkc1, and ROS as regulators of actin and suggest that ROS may play an important role in mTORC2-dependent dysregulation of the actin cytoskeleton in cancer cells.


Subject(s)
Actins/metabolism , Glycogen Synthase Kinase 3/metabolism , Multiprotein Complexes/metabolism , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Actin Cytoskeleton/metabolism , Blotting, Western , Cyclin C/genetics , Cyclin C/metabolism , Glycogen Synthase Kinase 3/genetics , Mechanistic Target of Rapamycin Complex 2 , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism , Mutation , Phosphorylation , Protein Binding , Protein Kinase C/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Subunits/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction , Sphingolipids/metabolism
14.
Proc Natl Acad Sci U S A ; 111(29): 10586-91, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25002487

ABSTRACT

The highly conserved Target of Rapamycin (TOR) kinase is a central regulator of cell growth and metabolism in response to nutrient availability. TOR functions in two structurally and functionally distinct complexes, TOR Complex 1 (TORC1) and TOR Complex 2 (TORC2). Through TORC1, TOR negatively regulates autophagy, a conserved process that functions in quality control and cellular homeostasis and, in this capacity, is part of an adaptive nutrient deprivation response. Here we demonstrate that during amino acid starvation TOR also operates independently as a positive regulator of autophagy through the conserved TORC2 and its downstream target protein kinase, Ypk1. Under these conditions, TORC2-Ypk1 signaling negatively regulates the Ca(2+)/calmodulin-dependent phosphatase, calcineurin, to enable the activation of the amino acid-sensing eIF2α kinase, Gcn2, and to promote autophagy. Our work reveals that the TORC2 pathway regulates autophagy in an opposing manner to TORC1 to provide a tunable response to cellular metabolic status.


Subject(s)
Amino Acids/pharmacology , Autophagy/drug effects , Glycogen Synthase Kinase 3/metabolism , Multiprotein Complexes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Calcineurin/metabolism , Mechanistic Target of Rapamycin Complex 2 , Models, Biological , Saccharomyces cerevisiae/cytology
15.
Cell Rep ; 6(3): 541-52, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24462291

ABSTRACT

Reactive oxygen species (ROS) are produced during normal metabolism and can function as signaling molecules. However, ROS at elevated levels can damage cells. Here, we identify the conserved target of rapamycin complex 2 (TORC2)/Ypk1 signaling module as an important regulator of ROS in the model eukaryotic organism, S. cerevisiae. We show that TORC2/Ypk1 suppresses ROS produced both by mitochondria as well as by nonmitochondrial sources, including changes in acidification of the vacuole. Furthermore, we link vacuole-related ROS to sphingolipids, essential components of cellular membranes, whose synthesis is also controlled by TORC2/Ypk1 signaling. In total, our data reveal that TORC2/Ypk1 act within a homeostatic feedback loop to maintain sphingolipid levels and that ROS are a critical regulatory signal within this system. Thus, ROS sensing and signaling by TORC2/Ypk1 play a central physiological role in sphingolipid biosynthesis and in the maintenance of cell growth and viability.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Homeostasis , Multiprotein Complexes/metabolism , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Signal Transduction , Sphingolipids/metabolism , TOR Serine-Threonine Kinases/metabolism , Acids/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Intracellular Space/metabolism , Mechanistic Target of Rapamycin Complex 2 , Microbial Viability , Mitochondria/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/growth & development , Vacuoles/metabolism
16.
Cell Cycle ; 11(20): 3745-9, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22895050

ABSTRACT

The PH domain-containing proteins Slm1 and Slm2 were originally identified as substrates of the rapamycin-insensitive TOR complex 2 (TORC2) and as mediators of signaling by the lipid second messenger phosphatidyl-inositol-4,5-bisphosphate (PI4,5P2) in budding yeast S. cerevisiae. More recently, these proteins have been identified as critical effectors that facilitate phosphorylation and activation of the AGC kinases Ypk1 and Ypk2 by TORC2. Here, we review the molecular basis for this regulation as well as place it within the context of recent findings that have revealed Slm1/2 and TORC2-dependent phosphorylation of Ypk1 is coupled to the biosynthesis of complex sphingolipids and to their levels within the plasma membrane (PM) as well as other forms of PM stress. Together, these studies reveal the existence of an intricate homeostatic feedback mechanism, whereby the activity of these signaling components is linked to the biosynthesis of PM lipids according to cellular need.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation, Fungal , Glycogen Synthase Kinase 3/genetics , Multiprotein Complexes/metabolism , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Sphingolipids/biosynthesis , TOR Serine-Threonine Kinases/metabolism , Carrier Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Cytoskeletal Proteins , Enzyme Activation , Feedback, Physiological , Glycogen Synthase Kinase 3/metabolism , Humans , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/genetics , Phosphorylation , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics
17.
ACS Chem Biol ; 7(6): 982-7, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22496512

ABSTRACT

The target of rapamycin (TOR) is a critical regulator of growth, survival, and energy metabolism. The allosteric TORC1 inhibitor rapamycin has been used extensively to elucidate the TOR related signal pathway but is limited by its inability to inhibit TORC2. We used an unbiased cell proliferation assay of a kinase inhibitor library to discover QL-IX-55 as a potent inhibitor of S. cerevisiae growth. The functional target of QL-IX-55 is the ATP-binding site of TOR2 as evidenced by the discovery of resistant alleles of TOR2 through rational design and unbiased selection strategies. QL-IX-55 is capable of potently inhibiting both TOR complex 1 and 2 (TORC1 and TORC2) as demonstrated by biochemical IP kinase assays (IC(50) <50 nM) and cellular assays for inhibition of substrate YPK1 phosphorylation. In contrast to rapamycin, QL-IX-55 is capable of inhibiting TORC2-dependent transcription, which suggests that this compound will be a powerful probe to dissect the Tor2/TORC2-related signaling pathway in yeast.


Subject(s)
Antifungal Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/drug effects , Sirolimus/pharmacology , Transcription Factors/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Antifungal Agents/chemistry , Cell Cycle Proteins/metabolism , Humans , Models, Molecular , Mycoses/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism
18.
Proc Natl Acad Sci U S A ; 109(5): 1536-41, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22307609

ABSTRACT

The yeast AGC kinase orthologs Ypk1 and Ypk2 control several important cellular processes, including actin polarization, endocytosis, and sphingolipid metabolism. Activation of Ypk1/2 requires phosphorylation by kinases localized at the plasma membrane (PM), including the 3-phosphoinositide-dependent kinase 1 orthologs Pkh1/Pkh2 and the target of rapamycin complex 2 (TORC2). Unlike their mammalian counterparts SGK and Akt, Ypk1 and Ypk2 lack an identifiable lipid-targeting motif; therefore, how these proteins are recruited to the PM has remained elusive. To explore Ypk1/2 function, we constructed ATP analog-sensitive alleles of both kinases and monitored global changes in gene expression following their inhibition, where we observed increased expression of stress-responsive target genes controlled by Ca(2+)-dependent phosphatase calcineurin. TORC2 has been shown previously to negatively regulate calcineurin in part by phosphorylating two related proteins, Slm1 and Slm2, which associate with the PM via plextrin homology domains. We therefore investigated the relationship between Slm1 and Ypk1 and discovered that these proteins interact physically and that Slm1 recruits Ypk1 to the PM for phosphorylation by TORC2. We observed further that these steps facilitate subsequent phosphorylation of Ypk1 by Pkh1/2. Remarkably, a requirement for Slm1, can be bypassed by fusing the plextrin homology domain of Slm1 alone onto Ypk1, demonstrating that the essential function of Slm1 is largely attributable to its role in Ypk1 activation. These findings both extend the scope of cellular processes regulated by Ypk1/2 to include negative regulation of calcineurin and broaden the repertoire of mechanisms for membrane recruitment and activation of a protein kinase.


Subject(s)
Carrier Proteins/physiology , Cell Cycle Proteins/physiology , Glycogen Synthase Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/physiology , RNA-Binding Proteins/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Cell Membrane/enzymology , Cytoskeletal Proteins , Enzyme Activation , Microscopy, Fluorescence
19.
Mol Cell ; 31(6): 775-6, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18922461

ABSTRACT

In a recent issue of Cell Metabolism, Porstmann et al. (2008) demonstrate that fatty acid biosynthesis, under the transcriptional control of SREBP1, is regulated by the rapamycin-sensitive mTOR signaling network, thus expanding the scope of biosynthetic processes integrated by mTOR.


Subject(s)
Fatty Acids/metabolism , Protein Kinases/metabolism , Animals , Cell Proliferation , Cholesterol/metabolism , Humans , Mice , Sterol Regulatory Element Binding Proteins/metabolism , TOR Serine-Threonine Kinases , Transcription Factors/metabolism
20.
Cell Metab ; 7(2): 148-58, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18249174

ABSTRACT

Ceramides and sphingoid long-chain bases (LCBs) are precursors to more complex sphingolipids and play distinct signaling roles crucial for cell growth and survival. Conserved reactions within the sphingolipid biosynthetic pathway are responsible for the formation of these intermediates. Components of target of rapamycin complex 2 (TORC2) have been implicated in the biosynthesis of sphingolipids in S. cerevisiae; however, the precise step regulated by this complex remains unknown. Here we demonstrate that yeast cells deficient in TORC2 activity are impaired for de novo ceramide biosynthesis both in vivo and in vitro. We find that TORC2 regulates this step in part by activating the AGC kinase Ypk2 and that this step is antagonized by the Ca2+/calmodulin-dependent phosphatase calcineurin. Because Ypk2 is activated independently by LCBs, the direct precursors to ceramides, our data suggest a model wherein TORC2 signaling is coupled with LCB levels to control Ypk2 activity and, ultimately, regulate ceramide formation.


Subject(s)
Cell Cycle Proteins/physiology , Ceramides/biosynthesis , Phosphatidylinositol 3-Kinases/physiology , Protein Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Fungal Proteins/metabolism , Oxidoreductases/metabolism , Signal Transduction , Yeasts
SELECTION OF CITATIONS
SEARCH DETAIL
...