Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35058357

ABSTRACT

After injury, severed dendrites and axons expose the "eat-me" signal phosphatidylserine (PS) on their surface while they break down. The degeneration of injured axons is controlled by a conserved Wallerian degeneration (WD) pathway, which is thought to activate neurite self-destruction through Sarm-mediated nicotinamide adenine dinucleotide (NAD+) depletion. While neurite PS exposure is known to be affected by genetic manipulations of NAD+, how the WD pathway coordinates both neurite PS exposure and self-destruction and whether PS-induced phagocytosis contributes to neurite breakdown in vivo remain unknown. Here, we show that in Drosophila sensory dendrites, PS exposure and self-destruction are two sequential steps of WD resulting from Sarm activation. Surprisingly, phagocytosis is the main driver of dendrite degeneration induced by both genetic NAD+ disruptions and injury. However, unlike neuronal Nmnat loss, which triggers PS exposure only and results in phagocytosis-dependent dendrite degeneration, injury activates both PS exposure and self-destruction as two redundant means of dendrite degeneration. Furthermore, the axon-death factor Axed is only partially required for self-destruction of injured dendrites, acting in parallel with PS-induced phagocytosis. Lastly, injured dendrites exhibit a unique rhythmic calcium-flashing that correlates with WD. Therefore, both NAD+-related general mechanisms and dendrite-specific programs govern PS exposure and self-destruction in injury-induced dendrite degeneration in vivo.


Subject(s)
Dendrites/metabolism , Phagocytosis , Sensory Receptor Cells/metabolism , Wallerian Degeneration/etiology , Wallerian Degeneration/metabolism , Animals , Drosophila , Drosophila Proteins/deficiency , Fluorescent Antibody Technique , Gene Knockdown Techniques , Nerve Degeneration , Nicotinamide-Nucleotide Adenylyltransferase/deficiency , Phosphatidylserines/metabolism , Wallerian Degeneration/pathology
2.
Elife ; 102021 12 08.
Article in English | MEDLINE | ID: mdl-34878972

ABSTRACT

Despite mounting evidence that the mammalian retina is exceptionally reliant on proper NAD+ homeostasis for health and function, the specific roles of subcellular NAD+ pools in retinal development, maintenance, and disease remain obscure. Here, we show that deletion of the nuclear-localized NAD+ synthase nicotinamide mononucleotide adenylyltransferase-1 (NMNAT1) in the developing murine retina causes early and severe degeneration of photoreceptors and select inner retinal neurons via multiple distinct cell death pathways. This severe phenotype is associated with disruptions to retinal central carbon metabolism, purine nucleotide synthesis, and amino acid pathways. Furthermore, transcriptomic and immunostaining approaches reveal dysregulation of a collection of photoreceptor and synapse-specific genes in NMNAT1 knockout retinas prior to detectable morphological or metabolic alterations. Collectively, our study reveals previously unrecognized complexity in NMNAT1-associated retinal degeneration and suggests a yet-undescribed role for NMNAT1 in gene regulation during photoreceptor terminal differentiation.


Subject(s)
Gene Deletion , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Photoreceptor Cells, Vertebrate/enzymology , Retinal Degeneration/enzymology , Retinal Neurons/enzymology , Animals , Female , Male , Mice , Nicotinamide-Nucleotide Adenylyltransferase/deficiency , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Neurons/pathology
3.
Neurochem Int ; 121: 86-97, 2018 12.
Article in English | MEDLINE | ID: mdl-30278188

ABSTRACT

Golgi fragmentation and loss of Nicotinamide Mononucleotide Adenylyltransferase 2 (NMNAT2) are the early key features of many neurodegenerative disorders. We investigated the link between NMNAT2 loss, Golgi fragmentation and axon degeneration. Golgi fragmentation in the cultured dorsal root ganglion (DRG) neurons resulted in caspase dependent axon degeneration and neuronal cell death. NMNAT2 depletion in the DRG neurons caused Golgi fragmentation and caspase dependent axon degeneration. NMNAT2 depletion did not cause ATP loss in the axons. These results indicate that NMNAT2 is required for maintenance of Golgi structure. Loss of Golgi structure or Nmnat2 depletion causes caspase dependent neurodegeneration. cytNmnat1 overexpression inhibited the axon degeneration induced by Golgi fragmentation or NMNAT2 depletion. These results also suggest that these degeneration signals converge on a common cytNmnat1 mediated axon protective program and are distinct from the SARM1 mediated caspase independent axon degeneration.


Subject(s)
Ganglia, Spinal/enzymology , Golgi Apparatus/enzymology , Neurons/enzymology , Nicotinamide-Nucleotide Adenylyltransferase/deficiency , Animals , Apoptosis/physiology , Cells, Cultured , Ganglia, Spinal/pathology , Golgi Apparatus/pathology , Mice , Neurons/pathology , Nicotinamide-Nucleotide Adenylyltransferase/antagonists & inhibitors , Nicotinamide-Nucleotide Adenylyltransferase/genetics , RNA, Small Interfering/administration & dosage
4.
Cell Rep ; 21(1): 10-16, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978465

ABSTRACT

Studies with the WldS mutant mouse have shown that axon and synapse pathology in several models of neurodegenerative diseases are mechanistically related to injury-induced axon degeneration (Wallerian degeneration). Crucially, an absence of SARM1 delays Wallerian degeneration as robustly as WldS, but their relative capacities to confer long-term protection against related, non-injury axonopathy and/or synaptopathy have not been directly compared. While Sarm1 deletion or WldS can rescue perinatal lethality and widespread Wallerian-like axonopathy in young NMNAT2-deficient mice, we report that an absence of SARM1 enables these mice to survive into old age with no overt phenotype, whereas those rescued by WldS invariantly develop a progressive neuromuscular defect in their hindlimbs from around 3 months of age. We therefore propose Sarm1 deletion as a more reliable tool than WldS for investigating Wallerian-like mechanisms in disease models and suggest that SARM1 blockade may have greater therapeutic potential than WLDS-related strategies.


Subject(s)
Armadillo Domain Proteins/genetics , Cytoskeletal Proteins/genetics , Genes, Lethal , Muscular Atrophy/genetics , Nerve Tissue Proteins/genetics , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Wallerian Degeneration/genetics , Animals , Armadillo Domain Proteins/antagonists & inhibitors , Armadillo Domain Proteins/deficiency , Axons/metabolism , Axons/pathology , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/deficiency , Disease Models, Animal , Female , Gene Deletion , Gene Expression Regulation , Hindlimb/innervation , Hindlimb/metabolism , Hindlimb/pathology , Humans , Locomotion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Atrophy/prevention & control , Nerve Tissue Proteins/deficiency , Nicotinamide-Nucleotide Adenylyltransferase/deficiency , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Time Factors , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology , Wallerian Degeneration/prevention & control
5.
Curr Biol ; 27(6): 784-794, 2017 Mar 20.
Article in English | MEDLINE | ID: mdl-28262487

ABSTRACT

Axons require the axonal NAD-synthesizing enzyme NMNAT2 to survive. Injury or genetically induced depletion of NMNAT2 triggers axonal degeneration or defective axon growth. We have previously proposed that axonal NMNAT2 primarily promotes axon survival by maintaining low levels of its substrate NMN rather than generating NAD; however, this is still debated. NMN deamidase, a bacterial enzyme, shares NMN-consuming activity with NMNAT2, but not NAD-synthesizing activity, and it delays axon degeneration in primary neuronal cultures. Here we show that NMN deamidase can also delay axon degeneration in zebrafish larvae and in transgenic mice. Like overexpressed NMNATs, NMN deamidase reduces NMN accumulation in injured mouse sciatic nerves and preserves some axons for up to three weeks, even when expressed at a low level. Remarkably, NMN deamidase also rescues axonal outgrowth and perinatal lethality in a dose-dependent manner in mice lacking NMNAT2. These data further support a pro-degenerative effect of accumulating NMN in axons in vivo. The NMN deamidase mouse will be an important tool to further probe the mechanisms underlying Wallerian degeneration and its prevention.


Subject(s)
Amidohydrolases/genetics , Axons/pathology , Nerve Degeneration/genetics , Nicotinamide-Nucleotide Adenylyltransferase/deficiency , Wallerian Degeneration/genetics , Amidohydrolases/metabolism , Animals , Mice , Mice, Transgenic , Nerve Degeneration/metabolism , Wallerian Degeneration/metabolism
6.
Cell Rep ; 17(3): 774-782, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27732853

ABSTRACT

Axon degeneration is a tightly regulated, self-destructive program that is a critical feature of many neurodegenerative diseases, but the molecular mechanisms regulating this program remain poorly understood. Here, we identify S-phase kinase-associated protein 1A (Skp1a), a core component of a Skp/Cullin/F-box (SCF)-type E3 ubiquitin ligase complex, as a critical regulator of axon degeneration after injury in mammalian neurons. Depletion of Skp1a prolongs survival of injured axons in vitro and in the optic nerve in vivo. We demonstrate that Skp1a regulates the protein level of the nicotinamide adenine dinucleotide (NAD)+ synthesizing enzyme nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2) in axons. Loss of axonal Nmnat2 contributes to a local ATP deficit that triggers axon degeneration. Knockdown of Skp1a elevates basal levels of axonal Nmnat2, thereby delaying axon degeneration through prolonged maintenance of axonal ATP. Consistent with Skp1a functioning through regulation of Nmnat2, Skp1a knockdown fails to protect axons from Nmnat2 knockdown. These results illuminate the molecular mechanism underlying Skp1a-dependent axonal destruction.


Subject(s)
Axons/enzymology , Axons/pathology , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Wallerian Degeneration/enzymology , Wallerian Degeneration/pathology , Animals , Cells, Cultured , Energy Metabolism , Mice , Nicotinamide-Nucleotide Adenylyltransferase/deficiency , Optic Nerve/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Sensory Receptor Cells/metabolism
7.
Cell Rep ; 10(12): 1974-81, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25818290

ABSTRACT

SARM1 function and nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) loss both promote axon degeneration, but their relative relationship in the process is unknown. Here, we show that NMNAT2 loss and resultant changes to NMNAT metabolites occur in injured SARM1-deficient axons despite their delayed degeneration and that axon degeneration specifically induced by NMNAT2 depletion requires SARM1. Strikingly, SARM1 deficiency also corrects axon outgrowth in mice lacking NMNAT2, independently of NMNAT metabolites, preventing perinatal lethality. Furthermore, NAMPT inhibition partially restores outgrowth of NMNAT2-deficient axons, suggesting that the NMNAT substrate, NMN, contributes to this phenotype. NMNAT2-depletion-dependent degeneration of established axons and restricted extension of developing axons are thus both SARM1 dependent, and SARM1 acts either downstream of NMNAT2 loss and NMN accumulation in a linear pathway or in a parallel branch of a convergent pathway. Understanding the pathway will help establish relationships with other modulators of axon survival and facilitate the development of effective therapies for axonopathies.


Subject(s)
Armadillo Domain Proteins/metabolism , Axons/metabolism , Cytoskeletal Proteins/metabolism , Nerve Degeneration/pathology , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Animals , Cell Survival/physiology , Mice , Nicotinamide-Nucleotide Adenylyltransferase/deficiency
8.
J Biol Chem ; 289(21): 14796-811, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24739386

ABSTRACT

NAD biosynthesis is of substantial interest because of its important roles in regulating various biological processes. Nicotinamide mononucleotide adenylyltransferase 3 (Nmnat3) is considered a mitochondria-localized NAD synthesis enzyme involved in de novo and salvage pathways. Although the biochemical properties of Nmnat3 are well documented, its physiological function in vivo remains unclear. In this study, we demonstrated that Nmnat3 was localized in the cytoplasm of mature erythrocytes and critically regulated their NAD pool. Deficiency of Nmnat3 in mice caused splenomegaly and hemolytic anemia, which was associated with the findings that Nmnat3-deficient erythrocytes had markedly lower ATP levels and shortened lifespans. However, the NAD level in other tissues were not apparently affected by the deficiency of Nmnat3. LC-MS/MS-based metabolomics revealed that the glycolysis pathway in Nmnat3-deficient erythrocytes was blocked at a glyceraldehyde 3-phosphate dehydrogenase (GAPDH) step because of the shortage of the coenzyme NAD. Stable isotope tracer analysis further demonstrated that deficiency of Nmnat3 resulted in glycolysis stall and a shift to the pentose phosphate pathway. Our findings indicate the critical roles of Nmnat3 in maintenance of the NAD pool in mature erythrocytes and the physiological impacts at its absence in mice.


Subject(s)
Anemia, Hemolytic/metabolism , Erythrocytes/metabolism , Glycolysis , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Adenosine Triphosphate/metabolism , Anemia, Hemolytic/genetics , Animals , Blotting, Western , Chromatography, Liquid , Cytoplasm/enzymology , Erythrocytes/ultrastructure , Metabolic Networks and Pathways/genetics , Metabolomics/methods , Mice , Mice, Knockout , Microscopy, Electron, Scanning , NAD/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/deficiency , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Splenomegaly/genetics , Splenomegaly/metabolism , Survival Analysis , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...