Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Physiol Rev ; 98(2): 813-880, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29488822

ABSTRACT

Neuronal cell death occurs extensively during development and pathology, where it is especially important because of the limited capacity of adult neurons to proliferate or be replaced. The concept of cell death used to be simple as there were just two or three types, so we just had to work out which type was involved in our particular pathology and then block it. However, we now know that there are at least a dozen ways for neurons to die, that blocking a particular mechanism of cell death may not prevent the cell from dying, and that non-neuronal cells also contribute to neuronal death. We review here the mechanisms of neuronal death by intrinsic and extrinsic apoptosis, oncosis, necroptosis, parthanatos, ferroptosis, sarmoptosis, autophagic cell death, autosis, autolysis, paraptosis, pyroptosis, phagoptosis, and mitochondrial permeability transition. We next explore the mechanisms of neuronal death during development, and those induced by axotomy, aberrant cell-cycle reentry, glutamate (excitoxicity and oxytosis), loss of connected neurons, aggregated proteins and the unfolded protein response, oxidants, inflammation, and microglia. We then reassess which forms of cell death occur in stroke and Alzheimer's disease, two of the most important pathologies involving neuronal cell death. We also discuss why it has been so difficult to pinpoint the type of neuronal death involved, if and why the mechanism of neuronal death matters, the molecular overlap and interplay between death subroutines, and the therapeutic implications of these multiple overlapping forms of neuronal death.


Subject(s)
Apoptosis/physiology , Cell Death/physiology , Microglia/metabolism , Neurons/metabolism , Animals , Humans , Phagocytosis/physiology , Signal Transduction/physiology
2.
Am J Pathol ; 194(7): 1374-1387, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38537932

ABSTRACT

Clear-cell renal cell carcinoma (ccRCC), a tubular epithelial malignancy, secretes tumor necrosis factor (TNF), which signals ccRCC cells in an autocrine manner via two cell surface receptors, TNFR1 and TNFR2, to activate shared and distinct signaling pathways. Selective ligation of TNFR2 drives cell cycle entry of malignant cells via a signaling pathway involving epithelial tyrosine kinase, vascular endothelial cell growth factor receptor type 2, phosphatidylinositol-3-kinase, Akt, pSer727-Stat3, and mammalian target of rapamycin. In this study, phosphorylated 4E binding protein-1 (4EBP1) serine 65 (pSer65-4EBP1) was identified as a downstream target of this TNFR2 signaling pathway. pSer65-4EBP1 expression was significantly elevated relative to total 4EBP1 in ccRCC tissue compared with that in normal kidneys, with signal intensity increasing with malignant grade. Selective ligation of TNFR2 with the TNFR2-specific mutein increased pSer65-4EBP1 expression in organ cultures that co-localized with internalized TNFR2 in mitochondria and increased expression of mitochondrially encoded COX (cytochrome c oxidase subunit) Cox1, as well as nuclear-encoded Cox4/5b subunits. Pharmacologic inhibition of mammalian target of rapamycin reduced both TNFR2-specific mutein-mediated phosphorylation of 4EBP1 and cell cycle activation in tumor cells while increasing cell death. These results signify the importance of pSer65-4EBP1 in mediating TNFR2-driven cell-cycle entry in tumor cells in ccRCC and implicate a novel relationship between the TNFR2/pSer65-4EBP1/COX axis and mitochondrial function.


Subject(s)
Adaptor Proteins, Signal Transducing , Carcinoma, Renal Cell , Cell Cycle Proteins , Cell Proliferation , Kidney Neoplasms , Mitochondria , Receptors, Tumor Necrosis Factor, Type II , Signal Transduction , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Neoplasms/genetics , Mitochondria/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Biosynthesis , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type II/genetics
3.
Proc Natl Acad Sci U S A ; 109(33): 13325-30, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22847423

ABSTRACT

RAS is frequently mutated in human cancers and has opposing effects on autophagy and tumorigenesis. Identifying determinants of the cellular responses to RAS is therefore vital in cancer research. Here, we show that autophagic activity dictates the cellular response to oncogenic RAS. N-terminal Apoptosis-stimulating of p53 protein 2 (ASPP2) mediates RAS-induced senescence and inhibits autophagy. Oncogenic RAS-expressing ASPP2((Δ3/Δ3)) mouse embryonic fibroblasts that escape senescence express a high level of ATG5/ATG12. Consistent with the notion that autophagy levels control the cellular response to oncogenic RAS, overexpressing ATG5, but not autophagy-deficient ATG5 mutant K130R, bypasses RAS-induced senescence, whereas ATG5 or ATG3 deficiency predisposes to it. Mechanistically, ASPP2 inhibits RAS-induced autophagy by competing with ATG16 to bind ATG5/ATG12 and preventing ATG16/ATG5/ATG12 formation. Hence, ASPP2 modulates oncogenic RAS-induced autophagic activity to dictate the cellular response to RAS: to proliferate or senesce.


Subject(s)
Autophagy , Fibroblasts/cytology , Fibroblasts/metabolism , Oncogenes , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Autophagy-Related Protein 12 , Autophagy-Related Protein 5 , Cellular Senescence , Embryo, Nonmammalian/cytology , Humans , Mice , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes/metabolism , Protein Binding , Protein Stability , Proteins/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism
5.
J Neurosci ; 33(28): 11329-45, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23843507

ABSTRACT

Axon regeneration after injury requires the extensive reconstruction, reorganization, and stabilization of the microtubule cytoskeleton in the growth cones. Here, we identify KIF3C as a key regulator of axonal growth and regeneration by controlling microtubule dynamics and organization in the growth cone. KIF3C is developmentally regulated. Rat embryonic sensory axons and growth cones contain undetectable levels of KIF3C protein that is locally translated immediately after injury. In adult neurons, KIF3C is axonally transported from the cell body and is enriched at the growth cone where it preferentially binds to tyrosinated microtubules. Functionally, the interaction of KIF3C with EB3 is necessary for its localization at the microtubule plus-ends in the growth cone. Depletion of KIF3C in adult neurons leads to an increase in stable, overgrown and looped microtubules because of a strong decrease in the microtubule frequency of catastrophes, suggesting that KIF3C functions as a microtubule-destabilizing factor. Adult axons lacking KIF3C, by RNA interference or KIF3C gene knock-out, display an impaired axonal outgrowth in vitro and a delayed regeneration after injury both in vitro and in vivo. Murine KIF3C knock-out embryonic axons grow normally but do not regenerate after injury because they are unable to locally translate KIF3C. These data show that KIF3C is an injury-specific kinesin that contributes to axon growth and regeneration by regulating and organizing the microtubule cytoskeleton in the growth cone.


Subject(s)
Axons/physiology , Kinesins/physiology , Microtubules/physiology , Nerve Regeneration/physiology , Animals , Cells, Cultured , Female , Growth Cones/metabolism , Growth Cones/physiology , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/pathology
6.
J Neurosci ; 33(46): 18175-89, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24227726

ABSTRACT

Intracellular tau aggregates are the neuropathological hallmark of several neurodegenerative diseases, including Alzheimer's disease, progressive supranuclear palsy, and cases of frontotemporal dementia, but the link between these aggregates and neurodegeneration remains unclear. Neuronal models recapitulating the main features of tau pathology are necessary to investigate the molecular mechanisms of tau malfunction, but current models show little and inconsistent spontaneous tau aggregation. We show that dorsal root ganglion (DRG) neurons in transgenic mice expressing human P301S tau (P301S-htau) develop tau pathology similar to that found in brain and spinal cord and a significant reduction in mechanosensation occurs before detectable fibrillar tau formation. DRG neuronal cultures established from adult P301S-htau mice at different ages retained the pattern of aberrant tau found in vivo. Moreover, htau became progressively hyperphosphorylated over 2 months in vitro beginning with nonsymptomatic neurons, while hyperphosphorylated P301S-htau-positive neurons from 5-month-old mice cultured for 2 months died preferentially. P301S-htau-positive neurons grew aberrant axons, including spheroids, typically found in human tauopathies. Neurons cultured at advanced stages of tau pathology showed a 60% decrease in the fraction of moving mitochondria. SEG28019, a novel O-GlcNAcase inhibitor, reduced steady-state pSer396/pSer404 phosphorylation over 7 weeks in a significant proportion of DRG neurons showing for the first time the possible beneficial effect of prolonged dosing of O-GlcNAcase inhibitor in vitro. Our system is unique in that fibrillar tau forms without external manipulation and provides an important new tool for understanding the mechanisms of tau dysfunction and for screening of compounds for treatment of tauopathies.


Subject(s)
Sensory Receptor Cells/metabolism , Tauopathies/metabolism , beta-N-Acetylhexosaminidases/antagonists & inhibitors , tau Proteins/biosynthesis , Animals , Cells, Cultured , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/pathology , Tauopathies/drug therapy , Tauopathies/genetics , Tauopathies/pathology , beta-N-Acetylhexosaminidases/metabolism , tau Proteins/genetics
7.
J Biol Chem ; 288(13): 9145-52, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23386613

ABSTRACT

Microglia are resident brain macrophages, which can cause neuronal loss when activated in infectious, ischemic, traumatic, and neurodegenerative diseases. Caspase-8 has both prodeath and prosurvival roles, mediating apoptosis and/or preventing RIPK1-mediated necroptosis depending on cell type and stimulus. We found that inflammatory stimuli (LPS, lipoteichoic acid, or TNF-α) caused an increase in caspase-8 IETDase activity in primary rat microglia without inducing apoptosis. Inhibition of caspase-8 with either Z-VAD-fmk or IETD-fmk resulted in necrosis of activated microglia. Inhibition of caspases with Z-VAD-fmk did not kill non-activated microglia, or astrocytes and neurons in any condition. Necrostatin-1, a specific inhibitor of RIPK1, prevented microglial caspase inhibition-induced death, indicating death was by necroptosis. In mixed cerebellar cultures of primary neurons, astrocytes, and microglia, LPS induced neuronal loss that was prevented by inhibition of caspase-8 (resulting in microglial necroptosis), and neuronal death was restored by rescue of microglia with necrostatin-1. We conclude that the activation of caspase-8 in inflamed microglia prevents their death by necroptosis, and thus, caspase-8 inhibitors may protect neurons in the inflamed brain by selectively killing activated microglia.


Subject(s)
Apoptosis , Caspase 8/chemistry , Caspase Inhibitors/pharmacology , Microglia/metabolism , Necrosis , Animals , Cell Survival , Cells, Cultured , Imidazoles/metabolism , Indoles/metabolism , Inflammation , Lipopolysaccharides/metabolism , Models, Biological , Neuroglia/metabolism , Neurons/metabolism , Rats , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
8.
J Neurosci ; 32(8): 2657-66, 2012 Feb 22.
Article in English | MEDLINE | ID: mdl-22357850

ABSTRACT

Milk-fat globule EGF factor-8 (MFG-E8, SED1, lactadherin) is known to mediate the phagocytic removal of apoptotic cells by bridging phosphatidylserine (PS)-exposing cells and the vitronectin receptor (VR) on phagocytes. However, we show here that MFG-E8 can mediate phagocytosis of viable neurons during neuroinflammation induced by lipopolysaccharide (LPS), thereby causing neuronal death. In vitro, inflammatory neuronal loss is independent of apoptotic pathways, and is inhibited by blocking the PS/MFG-E8/VR pathway (by adding PS blocking antibodies, annexin V, mutant MFG-E8 unable to bind VR, or VR antagonist). Neuronal loss is absent in Mfge8 knock-out cultures, but restored by adding recombinant MFG-E8, without affecting inflammation. In vivo, LPS-induced neuronal loss is reduced in the striatum of Mfge8 knock-out mice or by coinjection of an MFG-E8 receptor (VR) inhibitor into the rat striatum. Our data show that blocking MFG-E8-dependent phagocytosis preserves live neurons, implying that phagocytosis actively contributes to neuronal death during brain inflammation.


Subject(s)
Antigens, Surface/metabolism , Encephalitis/pathology , Milk Proteins/metabolism , Neurons/physiology , Phagocytosis/physiology , Amino Acid Chloromethyl Ketones/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Annexin A5/metabolism , Antigens, Surface/genetics , Cell Count , Cells, Cultured , Cerebellum/cytology , Coculture Techniques , Corpus Striatum/drug effects , Cytokines/metabolism , Disease Models, Animal , Encephalitis/chemically induced , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Etoposide/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , In Situ Nick-End Labeling , Integrin alphaVbeta3/metabolism , Lectins/metabolism , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Milk Proteins/genetics , Mutation/genetics , Neuroglia/physiology , Neurons/drug effects , Peptides, Cyclic/pharmacology , Phagocytosis/drug effects , Phagocytosis/genetics , Phosphatidylserines/pharmacology , Phosphopyruvate Hydratase/metabolism , Rats , Signal Transduction/drug effects , Signal Transduction/genetics , bcl-2-Associated X Protein/deficiency , beta-Galactosidase/metabolism
9.
Am J Pathol ; 180(4): 1454-64, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22330679

ABSTRACT

The expression of death receptor 3 (DR3), a member of the tumor necrosis factor (TNF) receptor superfamily, is up-regulated in human tubular epithelial cells (TECs) during renal injury, but its function in this setting remains unknown. We used cisplatin to induce renal injury in wild-type (DR3(+/+)) or congenitally deficient DR3(-/-) mice to examine the in vivo role of DR3. Cisplatin induced the expression of DR3, its ligand, TNF-like ligand 1A (TL1A), and TNF in TECs, as observed in human renal injury. Cisplatin increased apoptotic death of DR3(-/-) TECs by twofold compared with DR3(+/+) TECs, whereas it reduced the number of tubules expressing phospho-NF-κBp65(Ser276) by 50% at 72 hours. Similar degrees of induction of DR3, TL1A, and TNF, and changes in apoptosis and phospho-NF-κBp65(Ser276), were obtained in mouse kidney organ cultures treated with cisplatin for 3 hours, suggesting a direct effect on TECs. TNF was implicated in mediating cisplatin-induced tubular damage given that the in vivo co-administration of GM6001, an inhibitor of TNF maturation and release, significantly reduced TNF production and tubular damage. Moreover, TNF exacerbated, whereas TL1A reduced, cisplatin-induced apoptosis in the DR3(+/+) mouse proximal tubule cell line, TKPTS. Our data demonstrate that cisplatin-induced nephrotoxicity is mitigated by DR3 signaling, suggesting that this occurs by antagonizing pro-apoptotic signals induced by TNF. Therefore, activating DR3 may be beneficial in reducing acute kidney injury.


Subject(s)
Acute Kidney Injury/pathology , Receptors, Tumor Necrosis Factor, Member 25/physiology , Tumor Necrosis Factor-alpha/physiology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Animals , Apoptosis/drug effects , Cisplatin/pharmacology , Dipeptides/pharmacology , Drug Interactions , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Kidney Tubules/pathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Ligands , Mice , Mice, Mutant Strains , NF-kappa B/metabolism , Organ Culture Techniques , Phosphorylation/drug effects , Protease Inhibitors/pharmacology , Receptors, Tumor Necrosis Factor, Member 25/biosynthesis , Receptors, Tumor Necrosis Factor, Member 25/deficiency , Signal Transduction/physiology , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation/drug effects
10.
J Immunol ; 186(8): 4973-83, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21402900

ABSTRACT

It is well-known that dead and dying neurons are quickly removed through phagocytosis by the brain's macrophages, the microglia. Therefore, neuronal loss during brain inflammation has always been assumed to be due to phagocytosis of neurons subsequent to their apoptotic or necrotic death. However, we report in this article that under inflammatory conditions in primary rat cultures of neurons and glia, phagocytosis actively induces neuronal death. Specifically, two inflammatory bacterial ligands, lipoteichoic acid or LPS (agonists of glial TLR2 and TLR4, respectively), stimulated microglial proliferation, phagocytic activity, and engulfment of ∼30% of neurons within 3 d. Phagocytosis of neurons was dependent on the microglial release of soluble mediators (and peroxynitrite in particular), which induced neuronal exposure of the eat-me signal phosphatidylserine (PS). Surprisingly, however, eat-me signaling was reversible, so that blocking any step in a phagocytic pathway consisting of PS exposure, the PS-binding protein milk fat globule epidermal growth factor-8, and its microglial vitronectin receptor was sufficient to rescue up to 90% of neurons without reducing inflammation. Hence, our data indicate a novel form of inflammatory neurodegeneration, where inflammation can cause eat-me signal exposure by otherwise viable neurons, leading to their death through phagocytosis. Thus, blocking phagocytosis may prevent some forms of inflammatory neurodegeneration, and therefore might be beneficial during brain infection, trauma, ischemia, neurodegeneration, and aging.


Subject(s)
Apoptosis/immunology , Microglia/immunology , Neurons/immunology , Phagocytosis/immunology , Amyloid beta-Peptides/pharmacology , Animals , Antigens, Surface , Cells, Cultured , Cerebellum/cytology , Coculture Techniques , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Microglia/cytology , Microglia/metabolism , Milk Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Peptide Fragments/pharmacology , Phagocytosis/drug effects , Phosphatidylserines/pharmacology , Rats , Teichoic Acids/pharmacology , Time Factors , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
J Neurosci ; 30(7): 2623-35, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-20164347

ABSTRACT

Synaptic activity promotes resistance to diverse apoptotic insults, the mechanism behind which is incompletely understood. We show here that a coordinated downregulation of core components of the intrinsic apoptosis pathway by neuronal activity forms a key part of the underlying mechanism. Activity-dependent protection against apoptotic insults is associated with inhibition of cytochrome c release in most but not all neurons, indicative of anti-apoptotic signaling both upstream and downstream of this step. We find that enhanced firing activity suppresses expression of the proapoptotic BH3-only member gene Puma in a NMDA receptor-dependent, p53-independent manner. Puma expression is sufficient to induce cytochrome c loss and neuronal apoptosis. Puma deficiency protects neurons against apoptosis and also occludes the protective effect of synaptic activity, while blockade of physiological NMDA receptor activity in the developing mouse brain induces neuronal apoptosis that is preceded by upregulation of Puma. However, enhanced activity can also confer resistance to Puma-induced apoptosis, acting downstream of cytochrome c release. This mechanism is mediated by transcriptional suppression of apoptosome components Apaf-1 and procaspase-9, and limiting caspase-9 activity, since overexpression of procaspase-9 accelerates the rate of apoptosis in active neurons back to control levels. Synaptic activity does not exert further significant anti-apoptotic effects downstream of caspase-9 activation, since an inducible form of caspase-9 overrides the protective effect of synaptic activity, despite activity-induced transcriptional suppression of caspase-3. Thus, suppression of apoptotic gene expression may synergize with other activity-dependent events such as enhancement of antioxidant defenses to promote neuronal survival.


Subject(s)
Apoptosis/physiology , Neural Inhibition/physiology , Neurons/physiology , Signal Transduction/physiology , Synapses/physiology , 4-Aminopyridine/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Bicuculline/pharmacology , Caspase 9/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cytochromes c/metabolism , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Drug Combinations , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , GABA Antagonists/pharmacology , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Neural Inhibition/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Potassium Channel Blockers , Signal Transduction/drug effects , Staurosporine/pharmacology , Synapses/drug effects , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Time Factors , Transfection/methods , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/metabolism , Up-Regulation/drug effects
12.
J Neurosci ; 30(10): 3782-92, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20220013

ABSTRACT

Death receptor 3 is a proinflammatory member of the immunomodulatory tumor necrosis factor receptor superfamily, which has been implicated in several inflammatory diseases such as arthritis and inflammatory bowel disease. Intriguingly however, constitutive DR3 expression has been detected in the brains of mice, rats, and humans, although its neurological function remains unknown. By mapping the normal brain expression pattern of DR3, we found that DR3 is expressed specifically by cells of the neuron lineage in a developmentally regulated and region-specific pattern. Behavioral studies on DR3-deficient (DR3(ko)) mice showed that constitutive neuronal DR3 expression was required for stable motor control function in the aging adult. DR3(ko) mice progressively developed behavioral defects characterized by altered gait, dyskinesia, and hyperactivity, which were associated with elevated dopamine and lower serotonin levels in the striatum. Importantly, retrograde tracing showed that absence of DR3 expression led to the loss of corticostriatal innervation without significant neuronal loss in aged DR3(ko) mice. These studies indicate that DR3 plays a key nonredundant role in the retention of normal motor control function during aging in mice and implicate DR3 in progressive neurological disease.


Subject(s)
Aging/physiology , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Motor Skills/physiology , Receptors, Tumor Necrosis Factor, Member 25/physiology , Aging/genetics , Animals , Cell Communication/genetics , Cell Communication/physiology , Cerebral Cortex/growth & development , Cerebral Cortex/physiology , Corpus Striatum/growth & development , Corpus Striatum/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurotransmitter Agents/deficiency , Neurotransmitter Agents/genetics , Neurotransmitter Agents/physiology , Receptors, Tumor Necrosis Factor, Member 25/deficiency , Receptors, Tumor Necrosis Factor, Member 25/genetics
13.
Essays Biochem ; 65(7): 847-857, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34897457

ABSTRACT

How neurons die in neurodegenerative diseases is still unknown. The distinction between apoptosis as a genetically controlled mechanism, and necrosis, which was viewed as an unregulated process, has blurred with the ever-increasing number of necrotic-like death subroutines underpinned by genetically defined pathways. It is therefore pertinent to ask whether any of them apply to neuronal cell death in tauopathies. Although Alzheimer's disease (AD) is the most prevalent tauopathy, tauopathies comprise an array of over 30 diseases in which the cytoplasmic protein tau aggregates in neurons, and also, in some diseases, in glia. Animal models have sought to distil the contribution of tau aggregation to the cell death process but despite intensive research, no one mechanism of cell death has been unequivocally defined. The process of tau aggregation, and the fibrillar structures that form, touch on so many cellular functions that there is unlikely to be a simple linear pathway of death; as one is blocked another is likely to take the lead. It is timely to ask how far we have advanced into defining whether any of the molecular players in the new death subroutines participate in the death process. Here we briefly review the currently known cell death routines and explore what is known about their participation in tau aggregation-related cell death. We highlight the involvement of cell autonomous and the more recent non-cell autonomous pathways that may enhance tau-aggregate toxicity, and discuss recent findings that implicate microglial phagocytosis of live neurons with tau aggregates as a mechanism of death.


Subject(s)
Alzheimer Disease , Tauopathies , Alzheimer Disease/metabolism , Animals , Cell Death , Neurons/metabolism , Tauopathies/metabolism , tau Proteins/chemistry , tau Proteins/metabolism
14.
Sci Adv ; 7(43): eabg4980, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34669475

ABSTRACT

The microtubule-associated protein tau aggregates in multiple neurodegenerative diseases, causing inflammation and changing the inflammatory signature of microglia by unknown mechanisms. We have shown that microglia phagocytose live neurons containing tau aggregates cultured from P301S tau mice due to neuronal tau aggregate-induced exposure of the "eat me" signal phosphatidylserine. Here, we show that after phagocytosing tau aggregate-bearing neurons, microglia become hypophagocytic while releasing seed-competent insoluble tau aggregates. These microglia express a senescence-like phenotype, demonstrated by acidic ß-galactosidase activity, secretion of paracrine senescence-associated cytokines, and maturation of matrix remodeling enzymes, results that are corroborated in P301S mouse brains and ex vivo brain slices. In particular, the nuclear factor κB­dependent activation of matrix metalloprotease 3 (MMP3/stromelysin1) was replicated in brains from patients with tauopathy. These data show that microglia that have been activated to ingest live tau aggregates-bearing neurons behave hormetically, becoming hypofunctional while acting as vectors of tau aggregate spreading.

15.
ACS Chem Neurosci ; 12(11): 1885-1893, 2021 06 02.
Article in English | MEDLINE | ID: mdl-33689290

ABSTRACT

Aggregated tau protein is a core pathology present in several neurodegenerative diseases. Therefore, the development and application of positron emission tomography (PET) imaging radiotracers that selectively bind to aggregated tau in fibril form is of importance in furthering the understanding of these disorders. While radiotracers used in human PET studies offer invaluable insight, radiotracers that are also capable of visualizing tau fibrils in animal models are important tools for translational research into these diseases. Herein, we report the synthesis and characterization of a novel library of compounds based on the phenyl/pyridinylbutadienylbenzothiazoles/benzothiazolium (PBB3) backbone developed for this application. From this library, we selected the compound LM229, which binds to recombinant tau fibrils with high affinity (Kd = 3.6 nM) and detects with high specificity (a) pathological 4R tau aggregates in living cultured neurons and mouse brain sections from transgenic human P301S tau mice, (b) truncated human 151-351 3R (SHR24) and 4R (SHR72) tau aggregates in transgenic rat brain sections, and (c) tau neurofibrillary tangles in brain sections from Alzheimer's disease (3R/4R tau) and progressive supranuclear palsy (4R tau). With LM229 also shown to cross the blood-brain barrier in vivo and its effective radiolabeling with the radioisotope carbon-11, we have established a novel platform for PET translational studies using rodent transgenic tau models.


Subject(s)
Alzheimer Disease , tau Proteins , Alzheimer Disease/diagnostic imaging , Animals , Brain/diagnostic imaging , Brain/metabolism , Mice , Mice, Transgenic , Neurofibrillary Tangles/metabolism , Positron-Emission Tomography , Rats , Rats, Transgenic , tau Proteins/metabolism
16.
Biochim Biophys Acta ; 1793(9): 1508-15, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19289147

ABSTRACT

Concurrent mitochondrial elimination and autophagy in many systems has led to the proposal that autophagy is the main mechanism of mitochondrial turnover during development and under pathological conditions. The term mitophagy was coined to describe the selective removal of mitochondria by autophagy but the process itself is still contentious. Three questions are being debated: 1) Is there a specific removal of mitochondria by autophagy or is it non-selective or inadvertent? 2) What are the signals that drive this process? 3) Does removal of mitochondria increase or decrease cell viability? There is a mounting evidence for specific signals in/on mitochondria that drive mitochondrial removal from cells by autophagy. The process itself may be both selective and non-selective. In yeast, surprisingly, mitochondrial elimination occurs more by microautophagy (intracellular pinocytosis by the vacuolar membrane) than macroautophagy (initiated by stand-alone nascent double membrane structures known as autophagosomes). In mammalian cells, macroautophagy seems most prevalent though tools to study microautophagy are not well developed. Whilst lack of mitophagy seems to be deleterious, understanding the interplay between autophagy, mitochondrial performance, and cell pathology is a much-needed area of research.


Subject(s)
Autophagy , Mitochondria/pathology , Mitochondria/physiology , Animals , Humans , Saccharomyces cerevisiae/cytology
17.
J Neurochem ; 114(3): 772-83, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20477944

ABSTRACT

Puma (p53 up-regulated modulator of apoptosis) is a BH3-only protein member of the Bcl-2 family that controls apoptosis by regulating the release of pro-apoptotic factors from mitochondria. Previously, we reported that sodium arsenite (NaAsO(2)) induces Puma-dependent apoptosis in cortical neurons in a p53-independent manner. The following evidence shows that p53-independent Puma activation by NaAsO(2) is mediated by the p53-related protein TAp73: (i) NaAsO(2) causes TAp73alpha accumulation and increases p53-independent expression of p73 target genes; (ii) two p53 response elements in the Puma promoter are required for NaAsO(2)-mediated activation of a Puma reporter construct; (iii) expression of the inhibitory DeltaNp73alpha and DeltaNp73beta isoforms decreases NaAsO(2)-mediated induction of Puma and other p53-family target genes in a p53-null background; (iv) DeltaNp73alpha and DeltaNp73beta expression protects the neurons from NaAsO(2)-dependent apoptosis. Interestingly, although ER stressors also induce p53-independent, Puma-dependent apoptosis, they do not increase TAp73 expression while NaAsO(2) does not induce notable endoplasmic reticulum (ER) stress. In contrast, DNA damaging agents, okadaic acid, and H(2)O(2) all induce apoptosis in a strictly Puma- and p53-dependent manner. Hence, the pivotal position of Puma as mediator of apoptosis in cortical neurons is because of the availability of at least three independent signalling pathways that ensure its activation.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/physiology , Cerebral Cortex/physiology , Nerve Degeneration/metabolism , Neurons/cytology , Nuclear Proteins/physiology , Tumor Suppressor Proteins/physiology , Animals , Animals, Newborn , Apoptosis Regulatory Proteins/genetics , Cerebral Cortex/cytology , Mice , Mice, Knockout , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Neurons/physiology , Nuclear Proteins/genetics , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/genetics
19.
J Neurosci ; 28(53): 14401-15, 2008 Dec 31.
Article in English | MEDLINE | ID: mdl-19118173

ABSTRACT

Proteins containing extended polyglutamine repeats cause at least nine neurodegenerative disorders, but the mechanisms of disease-related neuronal death remain uncertain. We show that sympathetic neurons containing cytoplasmic inclusions formed by 97 glutamines expressed within human huntingtin exon1-enhanced green fluorescent protein (Q97) undergo a protracted form of nonapoptotic death that is insensitive to Bax deletion or caspase inhibition but is characterized by mitochondrial dysfunction. By treating the neurons with combined cytosine arabinoside and NGF withdrawal, we demonstrate that Q97 confers a powerful resistance to apoptosis at multiple levels: despite normal proapoptotic signaling (elevation of P-ser15-p53 and BimEL), there is no increase of Puma mRNA or Bax activation, both necessary for apoptosis. Even restoration of Bax translocation with overexpressed Puma does not activate apoptosis. We demonstrate that this robust inhibition of apoptosis is caused by Q97-mediated accumulation of Hsp70, which occurs through inhibition of proteasomal activity. Thus, apoptosis is reinstated by short hairpin RNA-mediated knockdown of Hsp70. These findings explain the rarity of apoptotic death in Q97-expressing neurons. Given the proteasomal blockade, we test whether enhancing lysosomal-mediated degradation with rapamycin reduces Q97 accumulation. Rapamycin reduces the amount of nonpathological Q25 by 70% over 3 d, but Q97 accumulation is unaffected. Interestingly, Q47 inclusions form more slowly as a result of constitutive lysosomal degradation, but faster-forming Q97 inclusions escape lysosomal control. Thus, cytoplasmic Q97 inclusions are refractory to clearance by proteasomal and lysosomal systems, leading to a toxicity that dominates over neuroprotective Hsp70. Our findings may explain the rarity of apoptosis but the inevitable cell death associated with polyQ inclusion diseases.


Subject(s)
Apoptosis/physiology , Exons/genetics , Inclusion Bodies/metabolism , Neurons/physiology , Peptides/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Superior Cervical Ganglion/cytology , Analysis of Variance , Animals , Animals, Newborn , Apoptosis/drug effects , Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/genetics , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Inclusion Bodies/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Nerve Growth Factor/pharmacology , Neurons/cytology , Peptides/genetics , Peptides/pharmacology , Rats , Rats, Wistar , Receptor, trkA/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Signal Transduction/drug effects , Time Factors , Transfection , bcl-2-Associated X Protein/metabolism
20.
Methods Mol Biol ; 527: 21-9, xi, 2009.
Article in English | MEDLINE | ID: mdl-19241002

ABSTRACT

Differential labelling techniques like differential in-gel electrophoresis (DIGE) enable mixing a control with an experimental sample prior to protein separation, thereby reducing complexity and greatly improving the resolution and analysis of changes in protein expression. Although the shift caused by phosphorylation to a more acidic pI can, in principle, reveal phosphorylation events using DIGE, analysis and verification of the phosphorylation are fraught with problems. Here we describe a differential phospho-labelling technique that obtains the same advantages as DIGE, which we named DIPPL, for differential phosphoprotein labelling. The technique involves labelling two samples, one with 32Pi (orthophosphate) and the other with 33Pi (orthophosphate). The two samples are mixed and proteins are separated on a single gel. Dried gels are exposed twice: once so that total radiation from 32P and 33P is collected on a film or screen; then acetate sheets are interposed between the gel and the screen such that 33P radiation is filtered out leaving 32P radiation to filter through. We demonstrate the utility of this approach by studying the MEK/ERK-dependent changes in stathmin phosphorylation induced by NGF in primary sympathetic neurons.


Subject(s)
Isotope Labeling/methods , Phosphoproteins/analysis , Phosphorus Radioisotopes/pharmacology , Animals , Electrophoresis, Gel, Two-Dimensional/methods , Humans , Phosphoproteins/metabolism , Phosphorus Radioisotopes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL