Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
PLoS Genet ; 18(6): e1010246, 2022 06.
Article in English | MEDLINE | ID: mdl-35737728

ABSTRACT

SARM1 is the founding member of the TIR-domain family of NAD+ hydrolases and the central executioner of pathological axon degeneration. SARM1-dependent degeneration requires NAD+ hydrolysis. Prior to the discovery that SARM1 is an enzyme, SARM1 was studied as a TIR-domain adaptor protein with non-degenerative signaling roles in innate immunity and invertebrate neurodevelopment, including at the Drosophila neuromuscular junction (NMJ). Here we explore whether the NADase activity of SARM1 also contributes to developmental signaling. We developed transgenic Drosophila lines that express SARM1 variants with normal, deficient, and enhanced NADase activity and tested their function in NMJ development. We find that NMJ overgrowth scales with the amount of NADase activity, suggesting an instructive role for NAD+ hydrolysis in this developmental signaling pathway. While degenerative and developmental SARM1 signaling share a requirement for NAD+ hydrolysis, we demonstrate that these signals use distinct upstream and downstream mechanisms. These results identify SARM1-dependent NAD+ hydrolysis as a heretofore unappreciated component of developmental signaling. SARM1 now joins sirtuins and Parps as enzymes that regulate signal transduction pathways via mechanisms that involve NAD+ cleavage, greatly expanding the potential scope of SARM1 TIR NADase functions.


Subject(s)
Armadillo Domain Proteins , NAD , Animals , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Axons/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Drosophila/genetics , Drosophila/metabolism , NAD/genetics , NAD+ Nucleosidase/genetics , NAD+ Nucleosidase/metabolism
2.
Mol Cell ; 82(9): 1643-1659.e10, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35334231

ABSTRACT

The NADase SARM1 (sterile alpha and TIR motif containing 1) is a key executioner of axon degeneration and a therapeutic target for several neurodegenerative conditions. We show that a potent SARM1 inhibitor undergoes base exchange with the nicotinamide moiety of nicotinamide adenine dinucleotide (NAD+) to produce the bona fide inhibitor 1AD. We report structures of SARM1 in complex with 1AD, NAD+ mimetics and the allosteric activator nicotinamide mononucleotide (NMN). NMN binding triggers reorientation of the armadillo repeat (ARM) domains, which disrupts ARM:TIR interactions and leads to formation of a two-stranded TIR domain assembly. The active site spans two molecules in these assemblies, explaining the requirement of TIR domain self-association for NADase activity and axon degeneration. Our results reveal the mechanisms of SARM1 activation and substrate binding, providing rational avenues for the design of new therapeutics targeting SARM1.


Subject(s)
Armadillo Domain Proteins , NAD , Armadillo Domain Proteins/genetics , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , NAD/metabolism , NAD+ Nucleosidase/metabolism , Protein Domains
3.
Mol Neurodegener ; 17(1): 1, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34991663

ABSTRACT

BACKGROUND: In response to injury, neurons activate a program of organized axon self-destruction initiated by the NAD+ hydrolase, SARM1. In healthy neurons SARM1 is autoinhibited, but single amino acid changes can abolish autoinhibition leading to constitutively active SARM1 enzymes that promote degeneration when expressed in cultured neurons. METHODS: To investigate whether naturally occurring human variants might disrupt SARM1 autoinhibition and potentially contribute to risk for neurodegenerative disease, we assayed the enzymatic activity of all 42 rare SARM1 alleles identified among 8507 amyotrophic lateral sclerosis (ALS) patients and 9671 controls. We then intrathecally injected mice with virus expressing SARM1 constructs to test the capacity of an ALS-associated constitutively active SARM1 variant to promote neurodegeneration in vivo. RESULTS: Twelve out of 42 SARM1 missense variants or small in-frame deletions assayed exhibit constitutive NADase activity, including more than half of those that are unique to the ALS patients or that occur in multiple patients. There is a > 5-fold enrichment of constitutively active variants among patients compared to controls. Expression of constitutively active ALS-associated SARM1 alleles in cultured dorsal root ganglion (DRG) neurons is pro-degenerative and cytotoxic. Intrathecal injection of an AAV expressing the common SARM1 reference allele is innocuous to mice, but a construct harboring SARM1V184G, the constitutively active variant found most frequently among the ALS patients, causes axon loss, motor dysfunction, and sustained neuroinflammation. CONCLUSIONS: These results implicate rare hypermorphic SARM1 alleles as candidate genetic risk factors for ALS and other neurodegenerative conditions.


Subject(s)
Amyotrophic Lateral Sclerosis , Armadillo Domain Proteins , Cytoskeletal Proteins , Neurodegenerative Diseases , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Armadillo Domain Proteins/chemistry , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Axons/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Humans , Mice , Neurodegenerative Diseases/metabolism
4.
Cell Rep ; 37(3): 109872, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34686345

ABSTRACT

SARM1 is an inducible TIR-domain NAD+ hydrolase that mediates pathological axon degeneration. SARM1 is activated by an increased ratio of NMN to NAD+, which competes for binding to an allosteric activating site. When NMN binds, the TIR domain is released from autoinhibition, activating its NAD+ hydrolase activity. The discovery of this allosteric activating site led us to hypothesize that other NAD+-related metabolites might activate SARM1. Here, we show the nicotinamide analog 3-acetylpyridine (3-AP), first identified as a neurotoxin in the 1940s, is converted to 3-APMN, which activates SARM1 and induces SARM1-dependent NAD+ depletion, axon degeneration, and neuronal death. In mice, systemic treatment with 3-AP causes rapid SARM1-dependent death, while local application to the peripheral nerve induces SARM1-dependent axon degeneration. We identify 2-aminopyridine as another SARM1-dependent neurotoxin. These findings identify SARM1 as a candidate mediator of environmental neurotoxicity and suggest that SARM1 agonists could be developed into selective agents for neurolytic therapy.


Subject(s)
Armadillo Domain Proteins/metabolism , Axons/drug effects , Cytoskeletal Proteins/metabolism , Ganglia, Spinal/drug effects , Nerve Degeneration , Neurotoxicity Syndromes/etiology , Neurotoxins/toxicity , Pyridines/toxicity , Sciatic Nerve/drug effects , Activation, Metabolic , Allosteric Regulation , Animals , Armadillo Domain Proteins/genetics , Axons/enzymology , Axons/pathology , Catalytic Domain , Cell Death , Cytokines/genetics , Cytokines/metabolism , Cytoskeletal Proteins/genetics , Enzyme Activation , Female , Ganglia, Spinal/enzymology , Ganglia, Spinal/pathology , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/pathology , Neurotoxins/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Pyridines/metabolism , Sciatic Nerve/enzymology , Sciatic Nerve/pathology , Signal Transduction
5.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: mdl-33468661

ABSTRACT

Axon degeneration is an active program of self-destruction mediated by the protein SARM1. In healthy neurons, SARM1 is autoinhibited and, upon injury autoinhibition is relieved, activating the SARM1 enzyme to deplete NAD+ and induce axon degeneration. SARM1 forms a homomultimeric octamer with each monomer composed of an N-terminal autoinhibitory ARM domain, tandem SAM domains that mediate multimerization, and a C-terminal TIR domain encoding the NADase enzyme. Here we discovered multiple intramolecular and intermolecular domain interfaces required for SARM1 autoinhibition using peptide mapping and cryo-electron microscopy (cryo-EM). We identified a candidate autoinhibitory region by screening a panel of peptides derived from the SARM1 ARM domain, identifying a peptide mediating high-affinity inhibition of the SARM1 NADase. Mutation of residues in full-length SARM1 within the region encompassed by the peptide led to loss of autoinhibition, rendering SARM1 constitutively active and inducing spontaneous NAD+ and axon loss. The cryo-EM structure of SARM1 revealed 1) a compact autoinhibited SARM1 octamer in which the TIR domains are isolated and prevented from oligomerization and enzymatic activation and 2) multiple candidate autoinhibitory interfaces among the domains. Mutational analysis demonstrated that five distinct interfaces are required for autoinhibition, including intramolecular and intermolecular ARM-SAM interfaces, an intermolecular ARM-ARM interface, and two ARM-TIR interfaces formed between a single TIR and two distinct ARM domains. These autoinhibitory regions are not redundant, as point mutants in each led to constitutively active SARM1. These studies define the structural basis for SARM1 autoinhibition and may enable the development of SARM1 inhibitors that stabilize the autoinhibited state.


Subject(s)
Armadillo Domain Proteins/chemistry , Cytoskeletal Proteins/chemistry , Ganglia, Spinal/enzymology , NAD/chemistry , Neurons/enzymology , Peptides/metabolism , Amino Acid Sequence , Animals , Armadillo Domain Proteins/antagonists & inhibitors , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Binding Sites , Cryoelectron Microscopy , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Ganglia, Spinal/cytology , Gene Expression , HEK293 Cells , Humans , Mice , Mice, Knockout , Models, Molecular , Mutation , NAD/metabolism , Neurons/cytology , Peptides/chemical synthesis , Primary Cell Culture , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
6.
Cell Rep ; 34(1): 108588, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33406435

ABSTRACT

Axonal degeneration is responsible for disease progression and accumulation of disability in many neurodegenerative conditions. The axonal degenerative process can generate a metastable pool of damaged axons that remain structurally and functionally viable but fated to degenerate in the absence of external intervention. SARM1, an NADase that depletes axonal energy stores upon activation, is the central driver of an evolutionarily conserved program of axonal degeneration. We identify a potent and selective small molecule isoquinoline inhibitor of SARM1 NADase that recapitulates the SARM1-/- phenotype and protects axons from degeneration induced by axotomy or mitochondrial dysfunction. SARM1 inhibition post-mitochondrial injury with rotenone allows recovery and rescues axons that already entered the metastable state. We conclude that SARM1 inhibition with small molecules has the potential to treat axonopathies of the central and peripheral nervous systems by preventing axonal degeneration and by allowing functional recovery of a metastable pool of damaged, but viable, axons.


Subject(s)
Armadillo Domain Proteins/drug effects , Armadillo Domain Proteins/physiology , Axons/physiology , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/physiology , Isoquinolines/pharmacology , Animals , Biomarkers/metabolism , Cell Line , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NAD+ Nucleosidase/drug effects , NAD+ Nucleosidase/physiology , Nerve Degeneration/drug therapy , Neurodegenerative Diseases/drug therapy , Phenotype , Recovery of Function
7.
J Exp Med ; 216(2): 294-303, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30642945

ABSTRACT

Axonal degeneration (AxD) following nerve injury, chemotherapy, and in several neurological disorders is an active process driven by SARM1, an injury-activated NADase. Axons of SARM1-null mice exhibit greatly delayed AxD after transection and in models of neurological disease, suggesting that inhibiting SARM1 is a promising strategy to reduce pathological AxD. Unfortunately, no drugs exist to target SARM1. We, therefore, developed SARM1 dominant-negatives that potently block AxD in cellular models of axotomy and neuropathy. To assess efficacy in vivo, we used adeno-associated virus-mediated expression of the most potent SARM1 dominant-negative and nerve transection as a model of severe AxD. While axons of vehicle-treated mice degenerate rapidly, axons of mice expressing SARM1 dominant-negative can remain intact for >10 d after transection, similar to the protection observed in SARM1-null mice. We thus developed a novel in vivo gene therapeutic to block pathological axon degeneration by inhibiting SARM1, an approach that may be applied clinically to treat manifold neurodegenerative diseases characterized by axon loss.


Subject(s)
Armadillo Domain Proteins , Axons/metabolism , Cytoskeletal Proteins , Dependovirus , Gene Targeting , Genetic Therapy , Nerve Degeneration , Animals , Armadillo Domain Proteins/antagonists & inhibitors , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Axons/pathology , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Mice , Mice, Transgenic , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/therapy , Transduction, Genetic
8.
Curr Biol ; 28(3): 421-430.e4, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29395922

ABSTRACT

The Toll/interleukin-1 receptor (TIR) domain is the signature signaling domain of Toll-like receptors (TLRs) and their adaptors, serving as a scaffold for the assembly of protein complexes for innate immune signaling [1, 2]. TIR domain proteins are also expressed in plants, where they mediate disease resistance [3, 4], and in bacteria, where they have been associated with virulence [5-9]. In pursuing our work on axon degeneration [10], we made the surprising discovery that the TIR domain of SARM1 (sterile alpha and TIR motif containing 1), a TLR adaptor protein, has enzymatic activity [11]. Upon axon injury, the SARM1 TIR domain cleaves nicotinamide adenine dinucleotide (NAD+), destroying this essential metabolic co-factor to trigger axon destruction [11, 12]. Whereas current studies of TIR domains focus on their scaffolding function, our findings with SARM1 inspired us to ask whether this enzymatic activity is the primordial function of the TIR domain. Here we show that ancestral prokaryotic TIR domains constitute a new family of NADase enzymes. Using purified proteins from a cell-free translation system, we find that TIR domain proteins from both bacteria and archaea cleave NAD+ into nicotinamide and ADP-ribose (ADPR), with catalytic cleavage executed by a conserved glutamic acid. A subset of bacterial and archaeal TIR domains generates a non-canonical variant cyclic ADPR (cADPR) molecule, and the full-length TIR domain protein from pathogenic Staphylococcus aureus induces NAD+ loss in mammalian cells. These findings suggest that the primordial function of the TIR domain is the enzymatic cleavage of NAD+ and establish TIR domain proteins as a new class of metabolic regulatory enzymes.


Subject(s)
Archaea/genetics , Archaeal Proteins/genetics , Bacteria/genetics , Bacterial Proteins/genetics , Animals , Archaea/enzymology , Archaeal Proteins/metabolism , Axons/metabolism , Bacteria/enzymology , Bacterial Proteins/metabolism , Mice
9.
Neuron ; 93(6): 1334-1343.e5, 2017 Mar 22.
Article in English | MEDLINE | ID: mdl-28334607

ABSTRACT

Axonal degeneration is an early and prominent feature of many neurological disorders. SARM1 is the central executioner of the axonal degeneration pathway that culminates in depletion of axonal NAD+, yet the identity of the underlying NAD+-depleting enzyme(s) is unknown. Here, in a series of experiments using purified proteins from mammalian cells, bacteria, and a cell-free protein translation system, we show that the SARM1-TIR domain itself has intrinsic NADase activity-cleaving NAD+ into ADP-ribose (ADPR), cyclic ADPR, and nicotinamide, with nicotinamide serving as a feedback inhibitor of the enzyme. Using traumatic and vincristine-induced injury models in neurons, we demonstrate that the NADase activity of full-length SARM1 is required in axons to promote axonal NAD+ depletion and axonal degeneration after injury. Hence, the SARM1 enzyme represents a novel therapeutic target for axonopathies. Moreover, the widely utilized TIR domain is a protein motif that can possess enzymatic activity.


Subject(s)
Armadillo Domain Proteins/metabolism , Axons/metabolism , Catalytic Domain , Cytoskeletal Proteins/metabolism , NAD+ Nucleosidase/metabolism , NAD/metabolism , Nerve Degeneration/metabolism , Animals , Armadillo Domain Proteins/genetics , Axons/pathology , Cells, Cultured , Cytoskeletal Proteins/genetics , Humans , Mice , Mice, Knockout , Nerve Degeneration/pathology
10.
Elife ; 52016 10 13.
Article in English | MEDLINE | ID: mdl-27735788

ABSTRACT

Overexpression of the NAD+ biosynthetic enzyme NMNAT1 leads to preservation of injured axons. While increased NAD+ or decreased NMN levels are thought to be critical to this process, the mechanism(s) of this axon protection remain obscure. Using steady-state and flux analysis of NAD+ metabolites in healthy and injured mouse dorsal root ganglion axons, we find that rather than altering NAD+ synthesis, NMNAT1 instead blocks the injury-induced, SARM1-dependent NAD+ consumption that is central to axon degeneration.


Subject(s)
Armadillo Domain Proteins/genetics , Axons/metabolism , Cytoskeletal Proteins/genetics , Nerve Degeneration/genetics , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Animals , Armadillo Domain Proteins/metabolism , Axons/pathology , Cytoskeletal Proteins/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Mice , NAD/metabolism , Nerve Degeneration/pathology
11.
J Neurochem ; 137(2): 154-63, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26778773

ABSTRACT

The control of NFκB in CNS neurons appears to differ from that in other cell types. Studies have reported induction of NFκB in neuronal cultures and immunostaining in vivo, but others have consistently detected little or no transcriptional activation by NFκB in brain neurons. To test if neurons lack some component of the signal transduction system for NFκB activation, we transfected cortical neurons with several members of this signaling system along with a luciferase-based NFκB-reporter plasmid; RelA was cotransfected in some conditions. No component of the NFκB pathway was permissive for endogenous NFκB activity, and none stimulated the activity of exogenous RelA. Surprisingly, however, the latter was inhibited by cotransfection of NFκB-inducing kinase (NIK). Fluorescence imaging of RelA indicated that co-expression of NIK sequestered RelA in the cytoplasm, similar to the effect of IκBα. NIK-knockout mice showed elevated expression of an NFκB-reporter construct in neurons in vivo. Cortical neurons cultured from NIK-knockout mice showed elevated expression of an NFκB-reporter transgene. Consistent with data from other cell types, a C-terminal fragment of NIK suppressed RelA activity in astrocytes as well as neurons. Therefore, the inhibitory ability of the NIK C-terminus was unbiased with regard to cell type. However, inhibition of NFκB by full-length NIK is a novel outcome that appears to be specific to CNS neurons. This has implications for unique aspects of transcription in the CNS, perhaps relevant to aspects of development, neuroplasticity, and neuroinflammation. Full-length NIK was found to inhibit (down arrow) transcriptional activation of NFκB in neurons, while it elevated (up arrow) activity in astrocytes. Deletion constructs corresponding to the N-terminus or C-terminus also inhibited NFκB in neurons, while only the C-terminus did so in astrocytes. One possible explanation is that the inhibition in neurons occurs via two different mechanisms, including the potential for a neuron-specific protein (e.g., one of the 14-3-3 class) to create a novel complex in neurons, whereas the C-terminus may interact directly with NFκB. [Structure of NIK is based on Liu J., Sudom A., Min X., Cao Z., Gao X., Ayres M., Lee F., Cao P., Johnstone S., Plotnikova O., Walker N., Chen G., and Wang Z. (2012) Structure of the nuclear factor κB-inducing kinase (NIK) kinase domain reveals a constitutively active conformation. J Biol Chem. 287, 27326-27334); N-terminal lobe is oriented at top].


Subject(s)
Central Nervous System/cytology , Gene Expression Regulation/genetics , NF-kappa B/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Embryo, Mammalian , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , NF-kappaB-Inducing Kinase
12.
J Exp Biol ; 216(Pt 12): 2283-92, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23531825

ABSTRACT

The reduction of protein synthesis has been associated with resistance to hypoxic cell death. Which components of the translation machinery control hypoxic sensitivity and the precise mechanism has not been systematically investigated, although a reduction in oxygen consumption has been widely assumed to be the mechanism. Using genetic reagents in Caenorhabditis elegans, we examined the effect on organismal survival after hypoxia of knockdown of 10 factors functioning at the three principal steps in translation. Reduction-of-function of all 10 translation factors significantly increased hypoxic survival to varying degrees, not fully accounted for by the level of translational suppression. Measurement of oxygen consumption showed that strong hypoxia resistance was possible without a significant decrease in oxygen consumption. Hypoxic sensitivity had no correlation with lifespan or reactive oxygen species sensitivity, two phenotypes associated with reduced translation. Resistance to tunicamycin, which produces misfolded protein toxicity, was the only phenotype that significantly correlated with hypoxic sensitivity. Translation factor knockdown was also hypoxia protective for mouse primary neurons. These data show that translation factor knockdown is hypoxia protective in both C. elegans and mouse neurons and that oxygen consumption does not necessarily determine survival; rather, mitigation of misfolded protein toxicity is more strongly associated with hypoxic protection.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/physiology , Eukaryotic Initiation Factors/genetics , Neurons/physiology , Oxygen Consumption , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Hypoxia , Cells, Cultured , Eukaryotic Initiation Factors/metabolism , Gene Knockdown Techniques , Hippocampus/physiology , Lentivirus/genetics , Mice , Polymerase Chain Reaction , RNA Interference , Transfection , Tunicamycin/metabolism
13.
Mol Cell Biol ; 30(21): 5033-42, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20733002

ABSTRACT

Prolonged cellular hypoxia results in energy failure and ultimately cell death. However, less-severe hypoxia can induce a cytoprotective response termed hypoxic preconditioning (HP). The unfolded protein response pathway (UPR) has been known for some time to respond to hypoxia and regulate hypoxic sensitivity; however, the role of the UPR, if any, in HP essentially has been unexplored. We have shown previously that a sublethal hypoxic exposure of the nematode Caenorhabditis elegans induces a protein chaperone component of the UPR (L. L. Anderson, X. Mao, B. A. Scott, and C. M. Crowder, Science 323:630-633, 2009). Here, we show that HP induces the UPR and that the pharmacological induction of misfolded proteins is itself sufficient to stimulate a delayed protective response to hypoxic injury that requires the UPR pathway proteins IRE-1, XBP-1, and ATF-6. HP also required IRE-1 but not XBP-1 or ATF-6; instead, GCN-2, which is known to suppress translation and induce an adaptive transcriptional response under conditions of UPR activation or amino acid deprivation, was required for HP. The phosphorylation of the translation factor eIF2α, an established mechanism of GCN-2-mediated translational suppression, was not necessary for HP. These data suggest a model where hypoxia-induced misfolded proteins trigger the activation of IRE-1, which along with GCN-2 controls an adaptive response that is essential to HP.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Hypoxia/metabolism , Protein Folding , Transcription Factors/chemistry , Transcription Factors/metabolism , Unfolded Protein Response , Animals , Animals, Genetically Modified , Base Sequence , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , DNA Primers/genetics , Genes, Helminth , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Ischemic Preconditioning , Models, Biological , Mutation , Protein Folding/drug effects , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factors/genetics , Tunicamycin/pharmacology , Unfolded Protein Response/drug effects
14.
J Neuroinflammation ; 6: 16, 2009 May 18.
Article in English | MEDLINE | ID: mdl-19450264

ABSTRACT

The unique physiology and function of neurons create differences in their cellular physiology, including their regulation of gene expression. We began several years ago exploring the relationships between the NFkappaB transcription factor, neuronal survival, and glutamate receptor activation in telencephalic neurons. These studies led us to conclude that this population of cells is nearly incapable of activating the NFkappaB that is nonetheless expressed at reasonable levels. A subset of the kappaB cis elements are instead bound by members of the Sp1 family in neurons. Also surprising was our discovery that Sp1 itself, typically described as ubiquitous, is severely restricted in expression within forebrain neurons; Sp4 seems to be substituted during neuronal differentiation. These findings and their implications for neuronal differentiation--as well as potential dedifferentiation during degenerative processes--are discussed here.


Subject(s)
Gene Expression Regulation , NF-kappa B/metabolism , Neurons/physiology , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Glutamic Acid/metabolism , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , NF-kappa B/genetics , Neurogenesis/genetics , Neurogenic Inflammation/genetics , Neurogenic Inflammation/physiopathology , Neurons/cytology , Prosencephalon/cytology , Prosencephalon/physiology , Signal Transduction , Sp1 Transcription Factor/genetics , Sp4 Transcription Factor/genetics , Sp4 Transcription Factor/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
15.
Science ; 323(5914): 630-3, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19179530

ABSTRACT

Hypoxia is important in a wide range of biological processes, such as animal hibernation and cell survival, and is particularly relevant in many diseases. The sensitivity of cells and organisms to hypoxic injury varies widely, but the molecular basis for this variation is incompletely understood. Using forward genetic screens in Caenorhabditis elegans, we isolated a hypoxia-resistant reduction-of-function mutant of rrt-1 that encodes an arginyl-transfer RNA (tRNA) synthetase, an enzyme essential for protein translation. Knockdown of rrt-1, and of most other genes encoding aminoacyl-tRNA synthetases, rescued animals from hypoxia-induced death, and the level of hypoxia resistance was inversely correlated with translation rate. The unfolded protein response was induced by hypoxia and was required for the hypoxia resistance of the reduction-of-function mutant of rrt-1. Thus, translational suppression produces hypoxia resistance, in part by reducing unfolded protein toxicity.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Arginine-tRNA Ligase/genetics , Arginine-tRNA Ligase/metabolism , Caenorhabditis elegans/physiology , Cell Hypoxia , Oxygen/physiology , Protein Biosynthesis , Amino Acid Sequence , Amino Acyl-tRNA Synthetases/genetics , Animals , Arginine-tRNA Ligase/chemistry , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/biosynthesis , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Longevity , Molecular Sequence Data , Muscle Cells/physiology , Mutation , Neurons/physiology , Oxygen Consumption , Protein Folding , RNA Interference , Transgenes
16.
Genetics ; 181(2): 483-96, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19047414

ABSTRACT

The sensitivity of an organism to hypoxic injury varies widely across species and among cell types. However, a systematic description of the determinants of metazoan hypoxic sensitivity is lacking. Toward this end, we screened a whole-genome RNAi library for genes that promote hypoxic sensitivity in Caenorhabditis elegans. RNAi knockdown of 198 genes conferred an invariant hypoxia-resistant phenotype (Hyp-r). Eighty-six per cent of these hyp genes had strong homologs in other organisms, 73 with human reciprocal orthologs. The hyp genes were distributed among multiple functional categories. Transcription factors, chromatin modifying enzymes, and intracellular signaling proteins were highly represented. RNAi knockdown of about half of the genes produced no apparent deleterious phenotypes. The hyp genes had significant overlap with previously identified life span extending genes. Testing of the RNAi's in a mutant background defective in somatic RNAi machinery showed that most genes function in somatic cells to control hypoxic sensitivity. DNA microarray analysis identified a subset of the hyp genes that may be hypoxia regulated. siRNA knockdown of human orthologs of the hyp genes conferred hypoxia resistance to transformed human cells for 40% of the genes tested, indicating extensive evolutionary conservation of the hypoxic regulatory activities. The results of the screen provide the first systematic picture of the genetic determinants of hypoxic sensitivity. The number and diversity of genes indicates a surprisingly nonredundant genetic network promoting hypoxic sensitivity.


Subject(s)
Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Cell Death/genetics , Cell Hypoxia/genetics , Genes, Helminth , Animals , Cell Line , Gene Expression , Humans , Phenotype , RNA Interference , RNA, Small Interfering/genetics , Species Specificity
17.
J Neurochem ; 100(5): 1300-14, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17316402

ABSTRACT

Sp-family transcription factors (Sp1, Sp3 and Sp4) contain a zinc-finger domain that binds to DNA sequences rich in G-C/T. As assayed by RT-PCR analysis of mRNA, western-blot analysis, immunofluorescence, and antibody-dependent "supershift" of DNA-binding assays, the prominent Sp-family factors in cerebral neurons were identified as Sp3 and Sp4. By contrast, glial cells were found to express Sp1 and Sp3. We previously showed that the pattern of G-C/T binding activity of Sp-family factors is rapidly and specifically altered by the calcium influx accompanying activation of glutamate receptors. Here, we demonstrate that Sp-factor activity is also lost after a cerebral ischemia/reperfusion injury in vivo. Consistent with its calcium-dependent nature, we found that glutamate's effect on Sp-family factors could be blocked by inhibitors of calpains, neutral cysteine proteases activated by calcium. Purified calpain I cleaved Sp3 and Sp4 into products that retained G-C/T-binding activity, consistent with species observed in glutamate-treated neurons. These data provide details of an impact of glutamate-receptor activation on molecular events connected to gene expression.


Subject(s)
Calpain/physiology , Excitatory Amino Acid Agonists/pharmacology , Neurons/metabolism , Receptors, Glutamate/physiology , Sp3 Transcription Factor/metabolism , Sp4 Transcription Factor/metabolism , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Glutamic Acid/pharmacology , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Sp1 Transcription Factor/metabolism
18.
J Biol Chem ; 281(47): 35863-72, 2006 Nov 24.
Article in English | MEDLINE | ID: mdl-17023425

ABSTRACT

In addition to their conventional G-C/T target sequences, Sp1 family transcription factors (Sp-factors) can interact with a subset of the target sequences for NFkappaB. Due to the low level of bona fide NFkappaB activity in most resting cells, this interaction between Sp-factors and kappaB-sites could play important roles in cell function. Here we used mutagenesis of a canonical kappaB element from the immunoglobulin and HIV promoters to identify the GC-rich sequences at each end required for Sp-factor targeting. Through screening of multiple kappaB elements, a sequence element located in the second intron of superoxide dismutase-2 (SOD2) was identified as a good candidate for both NFkappaB and Sp-factor binding. In neurons, the prominent proteins interacting with this site were Sp3 and Sp4, whereas Sp1, Sp3, and NFkappaB were associated with this site in astroglia. The neuronal Sp-factors repressed transcriptional activity through this kappaB-site. In contrast, astroglial Sp-factors activated promoter activity through the same element. NFkappaB contributed to control of the SOD2 kappaB element only in astrocytes. These findings imply that cell-type specificity of transcription in the central nervous system, particularly with regard to kappaB elements, may include two unique aspects of neurons: 1) a recalcitrant NFkappaB and 2) the substitution of Sp4 for Sp1.


Subject(s)
Astrocytes/metabolism , Gene Expression Regulation, Developmental , NF-kappa B/genetics , Neurons/metabolism , Superoxide Dismutase/genetics , Transcription, Genetic , Animals , Brain/embryology , Cell Nucleus/metabolism , Central Nervous System/metabolism , HIV/genetics , Immunoglobulins/genetics , NF-kappa B/chemistry , Protein Binding , Rats , Sp Transcription Factors/metabolism , Superoxide Dismutase/chemistry
19.
J Neurochem ; 97(3): 607-18, 2006 May.
Article in English | MEDLINE | ID: mdl-16573643

ABSTRACT

Nuclear factor kappaB (NFkappaB) is a dynamically modulated transcription factor with an extensive literature pertaining to widespread actions across species, cell types and developmental stages. Analysis of NFkappaB in a complex environment such as neural tissue suffers from a difficulty in simultaneously establishing both activity and location. Much of the available data indicate a profound recalcitrance of NFkappaB activation in neurons, as compared with most other cell types. Few studies to date have sought to distinguish between the various combinatorial dimers of NFkappaB family members. Recent research has illuminated the importance of these problems, as well as opportunities to move past them to the nuances manifest through variable activation pathways, subunit complexity and target sequence preferences.


Subject(s)
NF-kappa B/physiology , Neurobiology , Neurons/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Glutamic Acid/pharmacology , Humans , Models, Biological , Neurons/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
20.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 34(5): 412-6, 2005 09.
Article in Chinese | MEDLINE | ID: mdl-16216051

ABSTRACT

OBJECTIVE: To obtain peptides binding specifically to Pisum sativum agglutinin (PSA) from a phage-displayed random peptide library. METHODS: (1) A phage-displayed random hexapeptide library was screened with PSA as target. (2) Dot blot was used to analyze the influence of the alpha-Met-D-mannoside on binding between PSA and phage-displayed peptides. (3) Three peptides (RMWSF, RYDYSY, LRLRQL) were selectively synthesized, and different concentrations were used to inhibit PSA and ConA binding to the HRP. RESULTS: The enrichment occurred obviously after three rounds of screening. The insert sequences of amino acids, displayed on 22 phage DNAs from the third round of screening, were divided into three groups. The binding of phage-displayed peptides to PSA was specific as shown by dot blot and could be inhibited by alpha-Met-D-mannoside. LRLRQL was not dissolved in water. ARMWSF and RYDYSY inhibited binding of PSA to HRP, but failed to inhibit binding ConA to HRP. CONCLUSION: The binding site of peptides ARMWSF and RYDYSY is different to that of alpha-Met-D-mannoside.


Subject(s)
Peptides/metabolism , Plant Lectins/metabolism , Protein Binding , Binding Sites , Peptide Library , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...