Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 401
Filter
1.
J Clin Invest ; 134(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949024

ABSTRACT

Mitochondria-related neurodegenerative diseases have been implicated in the disruption of primary cilia function. Mutation in an intrinsic mitochondrial complex I component NDUFAF2 has been identified in Leigh syndrome, a severe inherited mitochondriopathy. Mutations in ARMC9, which encodes a basal body protein, cause Joubert syndrome, a ciliopathy with defects in the brain, kidney, and eye. Here, we report a mechanistic link between mitochondria metabolism and primary cilia signaling. We discovered that loss of NDUFAF2 caused both mitochondrial and ciliary defects in vitro and in vivo and identified NDUFAF2 as a binding partner for ARMC9. We also found that NDUFAF2 was both necessary and sufficient for cilia formation and that exogenous expression of NDUFAF2 rescued the ciliary and mitochondrial defects observed in cells from patients with known ARMC9 deficiency. NAD+ supplementation restored mitochondrial and ciliary dysfunction in ARMC9-deficient cells and zebrafish and ameliorated the ocular motility and motor deficits of a patient with ARMC9 deficiency. The present results provide a compelling mechanistic link, supported by evidence from human studies, between primary cilia and mitochondrial signaling. Importantly, our findings have significant implications for the development of therapeutic approaches targeting ciliopathies.


Subject(s)
Cilia , Kidney Diseases, Cystic , Leigh Disease , Mitochondria , Zebrafish , Humans , Zebrafish/metabolism , Zebrafish/genetics , Leigh Disease/genetics , Leigh Disease/metabolism , Leigh Disease/pathology , Cilia/metabolism , Cilia/pathology , Cilia/genetics , Animals , Mitochondria/metabolism , Mitochondria/pathology , Mitochondria/genetics , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/pathology , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Armadillo Domain Proteins/metabolism , Armadillo Domain Proteins/genetics , Retina/metabolism , Retina/pathology , Retina/abnormalities , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Eye Abnormalities/metabolism , Mice , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Cerebellum/metabolism , Cerebellum/pathology , Cerebellum/abnormalities , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Male
2.
J Cell Mol Med ; 28(12): e18449, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38924214

ABSTRACT

Mitochondrial dynamics has emerged as an important target for neuronal protection after cerebral ischaemia/reperfusion. Therefore, the aim of this study was to investigate the mechanism by which ARMC10 regulation of mitochondrial dynamics affects mitochondrial function involved in ischaemic stroke (IS). Mitochondrial morphology was detected by laser scanning confocal microscopy (LSCM), and mitochondrial ultrastructural alterations were detected by electron microscopy. The expression of mitochondrial dynamics-related genes Drp1, Mfn1, Mfn2, Fis1, OPA1 and ARMC10 and downstream target genes c-Myc, CyclinD1 and AXIN2 was detected by RT-qPCR. Western blot was used to detect the protein expression of ß-catenin, GSK-3ß, p-GSK-3ß, Bcl-2 and Bax. DCFH-DA fluorescent probe was to detect the effect of ARMC10 on mitochondrial ROS level, Annexin V-FITC fluorescent probe was to detect the effect of ARMC10 on apoptosis, and ATP assay kit was to detect the effect of ARMC10 on ATP production. Mitochondrial dynamics was dysregulated in clinical IS samples and in the OGD/R cell model, and the relative expression of ARMC10 gene was significantly decreased in IS group (p < 0.05). Knockdown and overexpression of ARMC10 could affect mitochondrial dynamics, mitochondrial function and neuronal apoptosis. Agonist and inhibitor affected mitochondrial function and neuronal apoptosis by targeting Wnt/ß-Catenin signal pathway. In the OGD/R model, ARMC10 affected mitochondrial function and neuronal apoptosis through the mechanism that regulates Wnt/ß-catenin signalling pathway. ARMC10 regulates mitochondrial dynamics and protects mitochondrial function by activating Wnt/ß-catenin signalling pathway, to exert neuroprotective effects.


Subject(s)
Apoptosis , Armadillo Domain Proteins , Ischemic Stroke , Mitochondria , Mitochondrial Dynamics , Wnt Signaling Pathway , Humans , Armadillo Domain Proteins/metabolism , Armadillo Domain Proteins/genetics , beta Catenin/metabolism , beta Catenin/genetics , Brain Ischemia/metabolism , Brain Ischemia/genetics , Brain Ischemia/pathology , Ischemic Stroke/metabolism , Ischemic Stroke/genetics , Ischemic Stroke/pathology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
3.
Cell Rep ; 43(6): 114362, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38870008

ABSTRACT

Wnt/Wingless (Wg) signaling is critical in development and disease, including cancer. Canonical Wnt signaling is mediated by ß-catenin/Armadillo (Arm in Drosophila) transducing signals to the nucleus, with IFT-A/Kinesin 2 complexes promoting nuclear translocation of ß-catenin/Arm. Here, we demonstrate that a conserved small N-terminal Arm34-87/ß-catenin peptide binds to IFT140, acting as a dominant interference tool to attenuate Wg/Wnt signaling in vivo. Arm34-87 expression antagonizes endogenous Wnt/Wg signaling, resulting in the reduction of its target expression. Arm34-87 inhibits Wg/Wnt signaling by interfering with nuclear translocation of endogenous Arm/ß-catenin, and this can be modulated by levels of wild-type ß-catenin or IFT140, with the Arm34-87 effect being enhanced or suppressed. Importantly, this mechanism is conserved in mammals with the equivalent ß-catenin24-79 peptide blocking nuclear translocation and pathway activation, including in cancer cells. Our work indicates that Wnt signaling can be regulated by a defined N-terminal ß-catenin peptide and thus might serve as an entry point for therapeutic applications to attenuate Wnt/ß-catenin signaling.


Subject(s)
Armadillo Domain Proteins , Cell Nucleus , Drosophila Proteins , Wnt Signaling Pathway , beta Catenin , beta Catenin/metabolism , Animals , Drosophila Proteins/metabolism , Cell Nucleus/metabolism , Humans , Armadillo Domain Proteins/metabolism , Armadillo Domain Proteins/genetics , Wnt1 Protein/metabolism , Wnt1 Protein/genetics , Active Transport, Cell Nucleus , Drosophila melanogaster/metabolism , Peptides/metabolism , Peptides/pharmacology , Protein Binding , Amino Acid Sequence , Transcription Factors
4.
Elife ; 122024 Jun 20.
Article in English | MEDLINE | ID: mdl-38900140

ABSTRACT

The Wnt/Wg pathway controls myriads of biological phenomena throughout the development and adult life of all organisms across the phyla. Thus, an aberrant Wnt signaling is associated with a wide range of pathologies in humans. Tight regulation of Wnt/Wg signaling is required to maintain proper cellular homeostasis. Here, we report a novel role of E3 ubiquitin ligase Deltex in Wg signaling regulation. Drosophila dx genetically interacts with wg and its pathway components. Furthermore, Dx LOF results in a reduced spreading of Wg while its over-expression expands the diffusion gradient of the morphogen. We attribute this change in Wg gradient to the endocytosis of Wg through Dx which directly affects the short- and long-range Wg targets. We also demonstrate the role of Dx in regulating Wg effector Armadillo where Dx down-regulates Arm through proteasomal degradation. We also showed the conservation of Dx function in the mammalian system where DTX1 is shown to bind with ß-catenin and facilitates its proteolytic degradation, spotlighting a novel step that potentially modulates Wnt/Wg signaling cascade.


Subject(s)
Armadillo Domain Proteins , Drosophila Proteins , Proteolysis , Ubiquitin-Protein Ligases , Wnt1 Protein , Animals , Humans , Armadillo Domain Proteins/metabolism , Armadillo Domain Proteins/genetics , beta Catenin/metabolism , beta Catenin/genetics , Drosophila/metabolism , Drosophila/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Signal Transduction , Transcription Factors , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Wnt Signaling Pathway , Wnt1 Protein/metabolism , Wnt1 Protein/genetics
5.
Proc Natl Acad Sci U S A ; 121(27): e2318198121, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38917007

ABSTRACT

Establishing modular binders as diagnostic detection agents represents a cost- and time-efficient alternative to the commonly used binders that are generated one molecule at a time. In contrast to these conventional approaches, a modular binder can be designed in silico from individual modules to, in principle, recognize any desired linear epitope without going through a selection and hit-validation process, given a set of preexisting, amino acid-specific modules. Designed armadillo repeat proteins (dArmRP) have been developed as modular binder scaffolds, and we report here the generation of highly specific dArmRP modules by yeast surface display selection, performed on a rationally designed dArmRP library. A selection strategy was developed to distinguish the binding difference resulting from a single amino acid mutation in the target peptide. Our reverse-competitor strategy introduced here employs the designated target as a competitor to increase the sensitivity when separating specific from cross-reactive binders that show similar affinities for the target peptide. With this switch in selection focus from affinity to specificity, we found that the enrichment during this specificity sort is indicative of the desired phenotype, regardless of the binder abundance. Hence, deep sequencing of the selection pools allows retrieval of phenotypic hits with only 0.1% abundance in the selectivity sort pool from the next-generation sequencing data alone. In a proof-of-principle study, a binder was created by replacing all corresponding wild-type modules with a newly selected module, yielding a binder with very high affinity for the designated target that has been successfully validated as a detection agent in western blot analysis.


Subject(s)
Armadillo Domain Proteins , Saccharomyces cerevisiae , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , High-Throughput Nucleotide Sequencing/methods , Protein Binding , Peptides/metabolism , Peptides/genetics , Peptides/chemistry , Epitopes/genetics , Peptide Library
6.
J Neurosci ; 44(24)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38692735

ABSTRACT

Sterile alpha and TIR motif containing 1 (SARM1) is an inducible NADase that localizes to mitochondria throughout neurons and senses metabolic changes that occur after injury. Minimal proteomic changes are observed upon either SARM1 depletion or activation, suggesting that SARM1 does not exert broad effects on neuronal protein homeostasis. However, whether SARM1 activation occurs throughout the neuron in response to injury and cell stress remains largely unknown. Using a semiautomated imaging pipeline and a custom-built deep learning scoring algorithm, we studied degeneration in both mixed-sex mouse primary cortical neurons and male human-induced pluripotent stem cell-derived cortical neurons in response to a number of different stressors. We show that SARM1 activation is differentially restricted to specific neuronal compartments depending on the stressor. Cortical neurons undergo SARM1-dependent axon degeneration after mechanical transection, and SARM1 activation is limited to the axonal compartment distal to the injury site. However, global SARM1 activation following vacor treatment causes both cell body and axon degeneration. Context-specific stressors, such as microtubule dysfunction and mitochondrial stress, induce axonal SARM1 activation leading to SARM1-dependent axon degeneration and SARM1-independent cell body death. Our data reveal that compartment-specific SARM1-mediated death signaling is dependent on the type of injury and cellular stressor.


Subject(s)
Armadillo Domain Proteins , Cerebral Cortex , Cytoskeletal Proteins , Induced Pluripotent Stem Cells , Neurons , Armadillo Domain Proteins/metabolism , Armadillo Domain Proteins/genetics , Animals , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Mice , Neurons/metabolism , Neurons/pathology , Male , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Humans , Female , Induced Pluripotent Stem Cells/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/metabolism , Nerve Degeneration/genetics , Cells, Cultured , Mice, Inbred C57BL , Stress, Physiological/physiology , Axons/metabolism , Axons/pathology , Mitochondria/metabolism
7.
Biomolecules ; 14(5)2024 May 07.
Article in English | MEDLINE | ID: mdl-38785968

ABSTRACT

Plakophilin 1 (PKP1), a member of the p120ctn subfamily of the armadillo (ARM)-repeat-containing proteins, is an important structural component of cell-cell adhesion scaffolds although it can also be ubiquitously found in the cytoplasm and the nucleus. RYBP (RING 1A and YY1 binding protein) is a multifunctional intrinsically disordered protein (IDP) best described as a transcriptional regulator. Both proteins are involved in the development and metastasis of several types of tumors. We studied the binding of the armadillo domain of PKP1 (ARM-PKP1) with RYBP by using in cellulo methods, namely immunofluorescence (IF) and proximity ligation assay (PLA), and in vitro biophysical techniques, namely fluorescence, far-ultraviolet (far-UV) circular dichroism (CD), and isothermal titration calorimetry (ITC). We also characterized the binding of the two proteins by using in silico experiments. Our results showed that there was binding in tumor and non-tumoral cell lines. Binding in vitro between the two proteins was also monitored and found to occur with a dissociation constant in the low micromolar range (~10 µM). Finally, in silico experiments provided additional information on the possible structure of the binding complex, especially on the binding ARM-PKP1 hot-spot. Our findings suggest that RYBP might be a rescuer of the high expression of PKP1 in tumors, where it could decrease the epithelial-mesenchymal transition in some cancer cells.


Subject(s)
Intrinsically Disordered Proteins , Plakophilins , Protein Binding , Humans , Plakophilins/metabolism , Plakophilins/genetics , Plakophilins/chemistry , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Armadillo Domain Proteins/metabolism , Armadillo Domain Proteins/chemistry , Armadillo Domain Proteins/genetics , Protein Domains , Circular Dichroism
8.
Int Immunopharmacol ; 134: 112193, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38723372

ABSTRACT

Retinal neurodegenerative diseases are a category of refractory blinding eye conditions closely associated with oxidative stress induced by mitochondrial dysfunction in retinal cells. SARM1, a core driver molecule leading to axonal degeneration, possesses NAD+ enzyme (NADase) activity. However, the role of the SARM1-NAD+ axis in oxidative stress-induced retinal cell death remains unclear. Here, we employed the SARM1 NADase inhibitor DSRM-3716 and established a glucose oxidase (GOx)-induced oxidative stress cell model. We found that compared to the GOx group, the DSRM-3716 pre-treated group reduced the hydrolysis of NAD+, inhibited the elevation of oxidative stress markers induced by GOx, decreased mitochondrial dysfunction, lowered the phosphorylation level of JNK, and attenuated the occurrence of pyroptosis in retinal and nerve cells, thereby providing protection for neurite growth. Further utilization of the JNK activator Anisomycin activated JNK, revealed that the JNK/c-Jun pathway down-regulated NMNAT2 expression. Consequently, it reduced cellular NAD+ synthesis, exacerbated mitochondrial dysfunction and cell pyroptosis, and reversed the protective effect of DSRM-3716 on cells. In summary, the inhibition of SARM1 NADase activity substantially mitigates oxidative damage to retinal cells and mitochondrial damage. Additionally, JNK simultaneously serves as both an upstream and downstream regulator in the SARM1-NAD+ axis, regulating retinal cell pyroptosis and neurite injury. Thus, this study provides new insights into the pathological processes of retinal cell oxidative stress and identifies potential therapeutic targets for retinal neurodegenerative diseases.


Subject(s)
Armadillo Domain Proteins , Cytoskeletal Proteins , NAD , Oxidative Stress , Armadillo Domain Proteins/metabolism , Armadillo Domain Proteins/genetics , Oxidative Stress/drug effects , Animals , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , NAD/metabolism , Retina/pathology , Retina/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Mice , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Cell Line , Pyroptosis/drug effects , Humans , NAD+ Nucleosidase/metabolism
9.
PLoS One ; 19(4): e0302251, 2024.
Article in English | MEDLINE | ID: mdl-38635746

ABSTRACT

Sterile alpha and TIR motif-containing 1 (SARM1) is a protein involved in programmed death of injured axons. Following axon injury or a drug-induced insult, the TIR domain of SARM1 degrades the essential molecule nicotinamide adenine dinucleotide (NAD+), leading to a form of axonal death called Wallerian degeneration. Degradation of NAD+ by SARM1 is essential for the Wallerian degeneration process, but accumulating evidence suggest that other activities of SARM1, beyond the mere degradation of NAD+, may be necessary for programmed axonal death. In this study we show that the TIR domains of both human and fruit fly SARM1 produce 1''-2' and 1''-3' glycocyclic ADP-ribose (gcADPR) molecules as minor products. As previously reported, we observed that SARM1 TIR domains mostly convert NAD+ to ADPR (for human SARM1) or cADPR (in the case of SARM1 from Drosophila melanogaster). However, we now show that human and Drosophila SARM1 additionally convert ~0.1-0.5% of NAD+ into gcADPR molecules. We find that SARM1 TIR domains produce gcADPR molecules both when purified in vitro and when expressed in bacterial cells. Given that gcADPR is a second messenger involved in programmed cell death in bacteria and likely in plants, we propose that gcADPR may play a role in SARM1-induced programmed axonal death in animals.


Subject(s)
NAD , Wallerian Degeneration , Animals , Humans , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology , NAD/metabolism , Drosophila melanogaster/metabolism , Axons/metabolism , Bacteria/metabolism , Adenosine Diphosphate Ribose/metabolism , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism
10.
Eye (Lond) ; 38(10): 1802-1809, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38538779

ABSTRACT

Programmed axon death is a druggable pathway of axon degeneration that has garnered considerable interest from pharmaceutical companies as a promising therapeutic target for various neurodegenerative disorders. In this review, we highlight mechanisms through which this pathway is activated in the retina and optic nerve, and discuss its potential significance for developing therapies for eye disorders and beyond. At the core of programmed axon death are two enzymes, NMNAT2 and SARM1, with pivotal roles in NAD metabolism. Extensive preclinical data in disease models consistently demonstrate remarkable, and in some instances, complete and enduring neuroprotection when this mechanism is targeted. Findings from animal studies are now being substantiated by genetic human data, propelling the field rapidly toward clinical translation. As we approach the clinical phase, the selection of suitable disorders for initial clinical trials targeting programmed axon death becomes crucial for their success. We delve into the multifaceted roles of programmed axon death and NAD metabolism in retinal and optic nerve disorders. We discuss the role of SARM1 beyond axon degeneration, including its potential involvement in neuronal soma death and photoreceptor degeneration. We also discuss genetic human data and environmental triggers of programmed axon death. Lastly, we touch upon potential therapeutic approaches targeting NMNATs and SARM1, as well as the nicotinamide trials for glaucoma. The extensive literature linking programmed axon death to eye disorders, along with the eye's suitability for drug delivery and visual assessments, makes retinal and optic nerve disorders strong contenders for early clinical trials targeting programmed axon death.


Subject(s)
Armadillo Domain Proteins , Axons , Nicotinamide-Nucleotide Adenylyltransferase , Optic Nerve Diseases , Humans , Optic Nerve Diseases/drug therapy , Optic Nerve Diseases/physiopathology , Optic Nerve Diseases/metabolism , Axons/physiology , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Animals , Armadillo Domain Proteins/metabolism , Cytoskeletal Proteins/metabolism , Apoptosis/physiology , Retinal Diseases/drug therapy , Retinal Diseases/metabolism , Retinal Diseases/physiopathology , NAD/metabolism
11.
Neuroscience ; 545: 111-124, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38492796

ABSTRACT

Armcx1 is a member of the ARMadillo repeat-Containing protein on the X chromosome (ARMCX) family, which is recognized to have evolutionary conserved roles in regulating mitochondrial transport and dynamics. Previous research has shown that Armcx1 is expressed at higher levels in mice after axotomy and in adult retinal ganglion cells after crush injury, and this protein increases neuronal survival and axonal regeneration. However, its role in traumatic brain injury (TBI) is unclear. Therefore, the aim of this study was to assess the expression of Armcx1 after TBI and to explore possible related mechanisms by which Armcx1 is involved in TBI. We used C57BL/6 male mice to model TBI and evaluated the role of Armcx1 in TBI by transfecting mice with Armcx1 small interfering RNA (siRNA) to inhibit Armcx1 expression 24 h before TBI modeling. Western blotting, immunofluorescence, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining, Nissl staining, transmission electron microscopy, adenosine triphosphate (ATP) level measurement, neuronal apoptosis analysis, neurological function scoring and the Morris water maze were performed. The results demonstrated that Armcx1 protein expression was elevated after TBI and that the Armcx1 protein was localized in neurons and astroglial cells in cortical tissue surrounding the injury site. In addition, inhibition of Armcx1 expression further led to impaired mitochondrial transport, abnormal morphology, reduced ATP levels, aggravation of neuronal apoptosis and neurological dysfunction, and decrease Miro1 expression. In conclusion, our findings indicate that Armcx1 may exert neuroprotective effects by ameliorating neurological injury after TBI through a mitochondrial transport pathway involving Miro1.


Subject(s)
Armadillo Domain Proteins , Brain Injuries, Traumatic , Mice, Inbred C57BL , Mitochondria , rho GTP-Binding Proteins , Animals , Male , Mice , Adenosine Triphosphate/metabolism , Apoptosis/physiology , Armadillo Domain Proteins/metabolism , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Maze Learning/physiology , Mitochondria/metabolism , Neurons/metabolism , Neurons/pathology , rho GTP-Binding Proteins/metabolism
12.
Acta Neuropathol Commun ; 12(1): 23, 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38331947

ABSTRACT

Glaucoma is one of the leading causes of irreversible blindness worldwide and vision loss in the disease results from the deterioration of retinal ganglion cells (RGC) and their axons. Metabolic dysfunction of RGC plays a significant role in the onset and progression of the disease in both human patients and rodent models, highlighting the need to better define the mechanisms regulating cellular energy metabolism in glaucoma. This study sought to determine if Sarm1, a gene involved in axonal degeneration and NAD+ metabolism, contributes to glaucomatous RGC loss in a mouse model with chronic elevated intraocular pressure (IOP). Our data demonstrate that after 16 weeks of elevated IOP, Sarm1 knockout (KO) mice retain significantly more RGC than control animals. Sarm1 KO mice also performed significantly better when compared to control mice during optomotor testing, indicating that visual function is preserved in this group. Our findings also indicate that Sarm1 KO mice display mild ocular developmental abnormalities, including reduced optic nerve axon diameter and lower visual acuity than controls. Finally, we present data to indicate that SARM1 expression in the optic nerve is most prominently associated with oligodendrocytes. Taken together, these data suggest that attenuating Sarm1 activity through gene therapy, pharmacologic inhibition, or NAD+ supplementation, may be a novel therapeutic approach for patients with glaucoma.


Subject(s)
Glaucoma , Retinal Ganglion Cells , Humans , Mice , Animals , Retinal Ganglion Cells/metabolism , Intraocular Pressure , NAD/metabolism , Glaucoma/genetics , Optic Nerve/metabolism , Axons/metabolism , Mice, Knockout , Disease Models, Animal , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism
13.
J Biophotonics ; 17(3): e202300370, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38185916

ABSTRACT

Axonal degeneration is a key component of neurodegenerative diseases such as Huntington's disease (HD), Alzheimer's disease, and amyotrophic lateral sclerosis. Nicotinamide, an NAD+ precursor, has long since been implicated in axonal protection and reduction of degeneration. However, studies on nicotinamide (NAm) supplementation in humans indicate that NAm has no protective effect. Sterile alpha and toll/interleukin receptor motif-containing protein 1 (SARM1) regulates several cell responses to axonal damage and has been implicated in promoting neuronal degeneration. SARM1 inhibition seems to result in protection from neuronal degeneration while hydrogen peroxide has been implicated in oxidative stress and axonal degeneration. The effects of laser-induced axonal damage in wild-type and HD dorsal root ganglion cells treated with NAm, hydrogen peroxide (H2O2), and SARM1 inhibitor DSRM-3716 were investigated and the cell body width, axon width, axonal strength, and axon shrinkage post laser-induced injury were measured.


Subject(s)
Huntington Disease , Hydrogen Peroxide , Animals , Mice , Humans , Niacinamide , Mice, Knockout , Neurons/metabolism , Cytoskeletal Proteins/metabolism , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism
14.
J Biol Chem ; 300(2): 105620, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38176648

ABSTRACT

Sterile alpha and HEAT/armadillo motif-containing protein (SARM1) was recently described as a NAD+-consuming enzyme and has previously been shown to regulate immune responses in macrophages. Neuronal SARM1 is known to contribute to axon degeneration due to its NADase activity. However, how SARM1 affects macrophage metabolism has not been explored. Here, we show that macrophages from Sarm1-/- mice display elevated NAD+ concentrations and lower cyclic ADP-ribose, a known product of SARM1-dependent NAD+ catabolism. Further, SARM1-deficient macrophages showed an increase in the reserve capacity of oxidative phosphorylation and glycolysis compared to WT cells. Stimulation of macrophages to a proinflammatory state by lipopolysaccharide (LPS) revealed that SARM1 restricts the ability of macrophages to upregulate glycolysis and limits the expression of the proinflammatory gene interleukin (Il) 1b, but boosts expression of anti-inflammatory Il10. In contrast, we show macrophages lacking SARM1 induced to an anti-inflammatory state by IL-4 stimulation display increased oxidative phosphorylation and glycolysis, and reduced expression of the anti-inflammatory gene, Fizz1. Overall, these data show that SARM1 fine-tunes immune gene transcription in macrophages via consumption of NAD+ and altered macrophage metabolism.


Subject(s)
Armadillo Domain Proteins , Cytoskeletal Proteins , Neurons , Animals , Mice , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Axons/metabolism , Cyclic ADP-Ribose/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , NAD/metabolism , Neurons/metabolism
15.
Mol Neurodegener ; 19(1): 13, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38282024

ABSTRACT

BACKGROUND: Bioenergetic maladaptations and axonopathy are often found in the early stages of neurodegeneration. Nicotinamide adenine dinucleotide (NAD), an essential cofactor for energy metabolism, is mainly synthesized by Nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2) in CNS neurons. NMNAT2 mRNA levels are reduced in the brains of Alzheimer's, Parkinson's, and Huntington's disease. Here we addressed whether NMNAT2 is required for axonal health of cortical glutamatergic neurons, whose long-projecting axons are often vulnerable in neurodegenerative conditions. We also tested if NMNAT2 maintains axonal health by ensuring axonal ATP levels for axonal transport, critical for axonal function. METHODS: We generated mouse and cultured neuron models to determine the impact of NMNAT2 loss from cortical glutamatergic neurons on axonal transport, energetic metabolism, and morphological integrity. In addition, we determined if exogenous NAD supplementation or inhibiting a NAD hydrolase, sterile alpha and TIR motif-containing protein 1 (SARM1), prevented axonal deficits caused by NMNAT2 loss. This study used a combination of techniques, including genetics, molecular biology, immunohistochemistry, biochemistry, fluorescent time-lapse imaging, live imaging with optical sensors, and anti-sense oligos. RESULTS: We provide in vivo evidence that NMNAT2 in glutamatergic neurons is required for axonal survival. Using in vivo and in vitro studies, we demonstrate that NMNAT2 maintains the NAD-redox potential to provide "on-board" ATP via glycolysis to vesicular cargos in distal axons. Exogenous NAD+ supplementation to NMNAT2 KO neurons restores glycolysis and resumes fast axonal transport. Finally, we demonstrate both in vitro and in vivo that reducing the activity of SARM1, an NAD degradation enzyme, can reduce axonal transport deficits and suppress axon degeneration in NMNAT2 KO neurons. CONCLUSION: NMNAT2 ensures axonal health by maintaining NAD redox potential in distal axons to ensure efficient vesicular glycolysis required for fast axonal transport.


Subject(s)
Axonal Transport , NAD , Nicotinamide-Nucleotide Adenylyltransferase , Animals , Mice , Adenosine Triphosphate/metabolism , Armadillo Domain Proteins/metabolism , Axons/metabolism , Cytoskeletal Proteins/metabolism , Glycolysis , Homeostasis , NAD/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/metabolism
16.
J Biol Chem ; 300(2): 105630, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38199568

ABSTRACT

Sterile alpha and toll/interleukin receptor motif-containing 1 (SARM1) is a critical regulator of axon degeneration that acts through hydrolysis of NAD+ following injury. Recent work has defined the mechanisms underlying SARM1's catalytic activity and advanced our understanding of SARM1 function in axons, yet the role of SARM1 signaling in other compartments of neurons is still not well understood. Here, we show in cultured hippocampal neurons that endogenous SARM1 is present in axons, dendrites, and cell bodies and that direct activation of SARM1 by the neurotoxin Vacor causes not just axon degeneration, but degeneration of all neuronal compartments. In contrast to the axon degeneration pathway defined in dorsal root ganglia, SARM1-dependent hippocampal axon degeneration in vitro is not sensitive to inhibition of calpain proteases. Dendrite degeneration downstream of SARM1 in hippocampal neurons is dependent on calpain 2, a calpain protease isotype enriched in dendrites in this cell type. In summary, these data indicate SARM1 plays a critical role in neurodegeneration outside of axons and elucidates divergent pathways leading to degeneration in hippocampal axons and dendrites.


Subject(s)
Armadillo Domain Proteins , Cytoskeletal Proteins , Neurons , Animals , Mice , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Axons/metabolism , Calpain/metabolism , Cytoskeletal Proteins/metabolism , Dendrites/metabolism , Neurons/metabolism , Signal Transduction
17.
J Cereb Blood Flow Metab ; 44(2): 224-238, 2024 02.
Article in English | MEDLINE | ID: mdl-37898107

ABSTRACT

Ischemic stroke causes secondary neurodegeneration in the thalamus ipsilateral to the infarction site and impedes neurological recovery. Axonal degeneration of thalamocortical fibers and autophagy overactivation are involved in thalamic neurodegeneration after ischemic stroke. However, the molecular mechanisms underlying thalamic neurodegeneration remain unclear. Sterile /Armadillo/Toll-Interleukin receptor homology domain protein (SARM1) can induce Wallerian degeneration. Herein, we aimed to investigate the role of SARM1 in thalamic neurodegeneration and autophagy activation after photothrombotic infarction. Neurological deficits measured using modified neurological severity scores and adhesive-removal test were ameliorated in Sarm1-/- mice after photothrombotic infarction. Compared with wild-type mice, Sarm1-/- mice exhibited unaltered infarct volume; however, there were markedly reduced neuronal death and gliosis in the ipsilateral thalamus. In parallel, autophagy activation was attenuated in the thalamus of Sarm1-/- mice after cerebral infarction. Thalamic Sarm1 re-expression in Sarm1-/- mice increased thalamic neurodegeneration and promoted autophagy activation. Auotophagic inhibitor 3-methyladenine partially alleviated thalamic damage induced by SARM1. Moreover, autophagic initiation through rapamycin treatment aggravated post-stroke neuronal death and gliosis in Sarm1-/- mice. Taken together, SARM1 contributes to secondary thalamic neurodegeneration after cerebral infarction, at least partly through autophagy inhibition. SARM1 deficiency is a potential therapeutic strategy for secondary thalamic neurodegeneration and functional deficits after stroke.


Subject(s)
Ischemic Stroke , Stroke , Mice , Animals , Gliosis , Cerebral Infarction/metabolism , Stroke/metabolism , Ischemic Stroke/metabolism , Thalamus/metabolism , Axons/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism
18.
Mol Neurobiol ; 61(7): 4783-4803, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38135853

ABSTRACT

Subarachnoid hemorrhage (SAH) is a devastating disease associated with a high mortality and morbidity. Exosomes have been considered as a potential therapeutic target for SAH. However, the effect of exosomes in SAH remains to be elucidated. In this study, we focused on investigating the effect of plasma exosomal lncRNA TM7SF3-AU1 in white matter injury after SAH. The SAH model was established by means of endovascular perforation. Exosomes were extracted from rat plasma samples. The expression of RNAs in the exosomes was detected by the transcriptomic microarray. Differentially expressed circRNA, lncRNA, and mRNA were obtained. The ceRNA network showed that the lncRNA TM7SF3-AU1 and miR-702-3p were closely associated with SARM1. Knocking down TM7SF3-AU1 promoted the expression of miR-702-3p and suppressed the expression of SARM1, and knocking down TM7SF3-AU1 also attenuated white matter injury after SAH. In addition, knocking down TM7SF3-AU1 improved the neurological deficits in locomotion, anxiety, learning, memory, and electrophysiological activity after SAH. Mechanistically, TM7SF3-AU1 was able to absorb miR-702-3p, which directly bind the SARM1 mRNA. Furthermore, the white matter injury attenuated by knockdown of TM7SF3-AU1 was partially reversed by the miR-702-3p antagomir in SAH rats. Taken together, this study showed that TM7SF3-AU1 acts as a sponge for miR-702-3p, reducing the inhibitory effect of miR-702-3p on SARM1, resulting in increased SARM1 expression and thus leading to white matter injury after SAH. Our study provides new insights into exosome-associated white matter injury. It also highlights TM7SF3-AU1 as a potential therapeutic target for white matter injury after SAH.


Subject(s)
Cytoskeletal Proteins , Exosomes , MicroRNAs , RNA, Long Noncoding , Rats, Sprague-Dawley , Signal Transduction , Subarachnoid Hemorrhage , White Matter , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Exosomes/metabolism , Male , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/genetics , Subarachnoid Hemorrhage/pathology , Subarachnoid Hemorrhage/complications , White Matter/pathology , White Matter/metabolism , Armadillo Domain Proteins/metabolism , Armadillo Domain Proteins/genetics , Rats
19.
Nat Commun ; 14(1): 7823, 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-38016954

ABSTRACT

Ultra-tight binding is usually observed for proteins associating with rigidified molecules. Previously, we demonstrated that femtomolar binders derived from the Armadillo repeat proteins (ArmRPs) can be designed to interact very tightly with fully flexible peptides. Here we show for ArmRPs with four and seven sequence-identical internal repeats that the peptide-ArmRP complexes display conformational dynamics. These dynamics stem from transient breakages of individual protein-residue contacts that are unrelated to overall unbinding. The labile contacts involve electrostatic interactions. We speculate that these dynamics allow attaining very high binding affinities, since they reduce entropic losses. Importantly, only NMR techniques can pick up these local events by directly detecting conformational exchange processes without complications from changes in solvent entropy. Furthermore, we demonstrate that the interaction surface of the repeat protein regularizes upon peptide binding to become more compatible with the peptide geometry. These results provide novel design principles for ultra-tight binders.


Subject(s)
Carrier Proteins , Peptides , Carrier Proteins/metabolism , Peptides/chemistry , Proteins/metabolism , Armadillo Domain Proteins/metabolism , Entropy , Protein Binding , Protein Conformation
20.
J Biochem ; 174(6): 533-548, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37725528

ABSTRACT

Sterile alpha and Toll/interleukin receptor motif-containing protein 1 (SARM1) is a NAD+ hydrolase that plays a key role in axonal degeneration and neuronal cell death. We reported that c-Jun N-terminal kinase (JNK) activates SARM1 through phosphorylation at Ser-548. The importance of SARM1 phosphorylation in the pathological process of Parkinson's disease (PD) has not been determined. We thus conducted the present study by using rotenone (an inducer of PD-like pathology) and neurons derived from induced pluripotent stem cells (iPSCs) from healthy donors and a patient with familial PD PARK2 (FPD2). The results showed that compared to the healthy neurons, FPD2 neurons were more vulnerable to rotenone-induced stress and had higher levels of SARM1 phosphorylation. Similar cellular events were obtained when we used PARK2-knockdown neurons derived from healthy donor iPSCs. These events in both types of PD-model neurons were suppressed in neurons treated with JNK inhibitors, Ca2+-signal inhibitors, or by a SARM1-knockdown procedure. The degenerative events were enhanced in neurons overexpressing wild-type SARM1 and conversely suppressed in neurons overexpressing the SARM1-S548A mutant. We also detected elevated SARM1 phosphorylation in the midbrain of PD-model mice. The results indicate that phosphorylated SARM1 plays an important role in the pathological process of rotenone-induced neurodegeneration.


Subject(s)
Parkinson Disease , Rotenone , Humans , Animals , Mice , Rotenone/pharmacology , Rotenone/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Cell Death , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...