Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 31
Filtrer
1.
Natl Sci Rev ; 11(6): nwae182, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38962715

RÉSUMÉ

Accumulation of aggregated α-synuclein (α-syn) in Lewy bodies is the pathological hallmark of Parkinson's disease (PD). Genetic mutations in lipid metabolism are causative for a subset of patients with Parkinsonism. The role of α-syn's lipid interactions in its function and aggregation is recognized, yet the specific lipids involved and how lipid metabolism issues trigger α-syn aggregation and neurodegeneration remain unclear. Here, we found that α-syn shows a preference for binding to lysophospholipids (LPLs), particularly targeting lysophosphatidylcholine (LPC) without relying on electrostatic interactions. LPC is capable of maintaining α-syn in a compact conformation, significantly reducing its propensity to aggregate both in vitro and within cellular environments. Conversely, a reduction in the production of cellular LPLs is associated with an increase in α-syn accumulation. Our work underscores the critical role of LPLs in preserving the natural conformation of α-syn to inhibit improper aggregation, and establishes a potential connection between lipid metabolic dysfunction and α-syn aggregation in PD.

2.
Nat Cell Biol ; 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951706

RÉSUMÉ

α-Synuclein (α-Syn) aggregation is closely associated with Parkinson's disease neuropathology. Physiologically, α-Syn promotes synaptic vesicle (SV) clustering and soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex assembly. However, the underlying structural and molecular mechanisms are uncertain and it is not known whether this function affects the pathological aggregation of α-Syn. Here we show that the juxtamembrane region of vesicle-associated membrane protein 2 (VAMP2)-a component of the SNARE complex that resides on SVs-directly interacts with the carboxy-terminal region of α-Syn through charged residues to regulate α-Syn's function in clustering SVs and promoting SNARE complex assembly by inducing a multi-component condensed phase of SVs, α-Syn and other components. Moreover, VAMP2 binding protects α-Syn against forming aggregation-prone oligomers and fibrils in these condensates. Our results suggest a molecular mechanism that maintains α-Syn's function and prevents its pathological amyloid aggregation, the failure of which may lead to Parkinson's disease.

3.
bioRxiv ; 2024 Mar 06.
Article de Anglais | MEDLINE | ID: mdl-38496494

RÉSUMÉ

Post-translational modifications (PTMs) of α-synuclein (α-syn) such as acetylation and phosphorylation play important yet distinct roles in regulating α-syn conformation, membrane binding, and amyloid aggregation. However, how PTMs regulate α-syn function in presynaptic terminals remains unclear. Previously, we reported that α-syn clusters synaptic vesicles (SV) 1, and neutral phospholipid lysophosphatidylcholine (LPC) can mediate this clustering 2. Here, based on our previous findings, we further demonstrate that N-terminal acetylation, which occurs under physiological condition and is irreversible in mammalian cells, significantly enhances the functional activity of α-syn in clustering SVs. Mechanistic studies reveal that this enhancement is caused by the N-acetylation-promoted insertion of α-syn's N-terminus and increased intermolecular interactions on the LPC-containing membrane. Our work demonstrates that N-acetylation fine-tunes α-syn-LPC interaction for mediating α-syn's function in SV clustering.

4.
Circulation ; 2024 Feb 08.
Article de Anglais | MEDLINE | ID: mdl-38328928

RÉSUMÉ

BACKGROUND: Heart failure (HF), which is the terminal stage of many cardiovascular diseases, is associated with low survival rates and a severe financial burden. The mechanisms, especially the molecular mechanism combined with new theories, underlying the pathogenesis of HF remain elusive. We demonstrate that phosphorylation-regulated dynamic liquid-liquid phase separation of HIP-55 (hematopoietic progenitor kinase 1-interacting protein of 55 kDa) protects against HF. METHODS: Fluorescence recovery after photobleaching assay, differential interference contrast analysis, pull-down assay, immunofluorescence, and immunohistochemical analysis were used to investigate the liquid-liquid phase separation capacity of HIP-55 and its dynamic regulation in vivo and in vitro. Mice with genetic deletion of HIP-55 and mice with cardiac-specific overexpression of HIP-55 were used to examine the role of HIP-55 on ß-adrenergic receptor hyperactivation-induced HF. Mutation analysis and mice with specific phospho-resistant site mutagenesis were used to identify the role of phosphorylation-regulated dynamic liquid-liquid phase separation of HIP-55 in HF. RESULTS: Genetic deletion of HIP-55 aggravated HF, whereas cardiac-specific overexpression of HIP-55 significantly alleviated HF in vivo. HIP-55 possesses a strong capacity for phase separation. Phase separation of HIP-55 is dynamically regulated by AKT-mediated phosphorylation at S269 and T291 sites, failure of which leads to impairment of HIP-55 dynamic phase separation by formation of abnormal aggregation. Prolonged sympathetic hyperactivation stress induced decreased phosphorylation of HIP-55 S269 and T291, dysregulated phase separation, and subsequent aggregate formation of HIP55. Moreover, we demonstrated the important role of dynamic phase separation of HIP-55 in inhibiting hyperactivation of the ß-adrenergic receptor-mediated P38/MAPK (mitogen-activated protein kinase) signaling pathway. A phosphorylation-deficient HIP-55 mutation, which undergoes massive phase separation and forms insoluble aggregates, loses the protective activity against HF. CONCLUSIONS: Our work reveals that the phosphorylation-regulated dynamic phase separation of HIP-55 protects against sympathetic/adrenergic system-mediated heart failure.

5.
Proc Natl Acad Sci U S A ; 120(41): e2311416120, 2023 10 10.
Article de Anglais | MEDLINE | ID: mdl-37782781

RÉSUMÉ

An evolutionarily conserved region of the TDP-43 low-complexity domain (LCD) twenty residues in length can adopt either an α-helical or ß-strand conformation. When in the latter conformation, TDP-43 self-associates via the formation of a labile, cross-ß structure. Self-association can be monitored via the formation of phase-separated protein droplets. Exposure of droplets to hydrogen peroxide leads to oxidation of conserved methionine residues distributed throughout the LCD. Oxidation disassembles the cross-ß structure, thus eliminating both self-association and phase separation. Here, we demonstrate that this process reciprocally enables formation of α-helical structure in precisely the same region formerly functioning to facilitate ß-strand-mediated self-association. We further observe that the α-helical conformation allows interaction with a lipid-like detergent and that exposure to lipids enhances the ß-to-α conformational switch. We hypothesize that regulation of this oxidative switch will prove to be important to the control of localized translation within vertebrate cells. The experimental observations reported herein were heavily reliant on studies of 1,6-hexanediol, a chemical agent that selectively dissolves labile structures formed via the self-association of protein domains of low sequence complexity. This aliphatic alcohol is shown to exert its dissociative activity primarily via hydrogen-bonding interactions with carbonyl oxygen atoms of the polypeptide backbone. Such observations underscore the central importance of backbone-mediated protein:protein interactions that facilitate the self-association and phase separation of LCDs.


Sujet(s)
Protéines de liaison à l'ADN , Peptides , Protéines de liaison à l'ADN/métabolisme , Peptides/composition chimique , Domaines protéiques , Méthionine/métabolisme , Stress oxydatif
6.
bioRxiv ; 2023 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-37693418

RÉSUMÉ

An evolutionarily conserved region of the TDP-43 low complexity domain twenty residues in length can adopt either an α-helical or ß-strand conformation. When in the latter conformation, TDP-43 self-associates via the formation of a labile, cross-ß structure. Self-association can be monitored via the formation of phase separated protein droplets. Exposure of droplets to hydrogen peroxide leads to oxidation of conserved methionine residues distributed throughout the low complexity domain. Oxidation disassembles the cross-ß structure, thus eliminating both self-association and phase separation. Here we demonstrate that this process reciprocally enables formation of α-helical structure in precisely the same region formerly functioning to facilitate ß-strand mediated self-association. We further observe that the α-helical conformation allows interaction with a lipid-like detergent, and that exposure to lipids enhances the ß-to-α conformational switch. We hypothesize that regulation of this oxidative switch will prove to be important to the control of localized translation within vertebrate cells. The experimental observations reported herein were heavily reliant on studies of 1,6-hexanediol, a chemical agent that selectively dissolves labile structures formed via the self-association of protein domains of low sequence complexity. This aliphatic alcohol is shown to exert its dissociative activity primarily via hydrogen bonding interactions with carbonyl oxygen atoms of the polypeptide backbone. Such observations underscore the central importance of backbone-mediated protein:protein interactions that facilitate the self-association and phase separation of low complexity domains. Significance Statement: The TDP-43 protein is a constituent of RNA granules involved in regulated translation. TDP-43 contains a C-terminal domain of 150 amino acids of low sequence complexity conspicuously decorated with ten methionine residues. An evolutionarily conserved region (ECR) of 20 residues within this domain can adopt either of two forms of labile secondary structure. Under normal conditions wherein methionine residues are reduced, the ECR forms a labile cross-ß structure that enables RNA granule condensation. Upon methionine oxidation, the ECR undergoes a conformational switch to become an α-helix incompatible with self-association and granule integrity. Oxidation of the TDP-43 low complexity domain is hypothesized to occur proximal to mitochondria, thus facilitating dissolution of RNA granules and activation of localized translation.

7.
ACS Nano ; 17(11): 10129-10141, 2023 06 13.
Article de Anglais | MEDLINE | ID: mdl-37204199

RÉSUMÉ

Protein liquid-liquid phase separation (LLPS) plays a crucial role in mediating dynamic assembly of different membraneless organelles such as stress granules (SGs). Dysregulation of dynamic protein LLPS leads to aberrant phase transition and amyloid aggregation which is closely associated with neurodegenerative diseases. In this study, we found that three types of graphene quantum dots (GQDs) exhibit potent activity in preventing SG formation and promoting SG disassembly. We next demonstrate that GQDs can directly interact with the SGs-containing protein fused in sarcoma (FUS), inhibit and reverse FUS LLPS, and prevent its abnormal phase transition. Moreover, GQDs display superior activity in preventing amyloid aggregation of FUS and disaggregating preformed FUS fibrils. Mechanistic study further demonstrates that GQDs with different edge-site exhibit distinct binding affinity to FUS monomers and fibrils, which accounts for their distinct activities in modulating FUS LLPS and fibrillation. Our work reveals the potent capability of GQDs in modulating SG assembly, protein LLPS, and fibrillation and sheds light on rational design of GQDs as effective modulators of protein LLPS for therapeutics application.


Sujet(s)
Graphite , Boîtes quantiques , Sarcomes , Humains , Graphite/pharmacologie , Granules de stress , Amyloïde , Protéines amyloïdogènes , Transition de phase
8.
Methods Mol Biol ; 2563: 261-268, 2023.
Article de Anglais | MEDLINE | ID: mdl-36227478

RÉSUMÉ

Protein liquid-liquid phase separation (LLPS) plays an essential role in the dynamic assembly of various membraneless compartments, which fulfill different biological functions in cells. Numerous proteins were found to undergo LLPS in different conditions. However, a general approach to systemically identify and compare the LLPS ability of different proteins is lacking. Here, we introduce a high-throughput protein phase separation (HiPPS) profiling method to evaluate the LLPS ability of proteins using a combination of crystallization robot/manual mixing mode and high-content analysis system. This method enables us to rapidly and comprehensively explore the LLPS behavior of each individual protein as well as mixture of different proteins.


Sujet(s)
Protéines intrinsèquement désordonnées , Cristallisation , Protéines intrinsèquement désordonnées/composition chimique , Extraction liquide-liquide/méthodes
9.
Nat Cell Biol ; 24(9): 1378-1393, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-36075972

RÉSUMÉ

While acetylated, RNA-binding-deficient TDP-43 reversibly phase separates within nuclei into complex droplets (anisosomes) comprised of TDP-43-containing liquid outer shells and liquid centres of HSP70-family chaperones, cytoplasmic aggregates of TDP-43 are hallmarks of multiple neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Here we show that transient oxidative stress, proteasome inhibition or inhibition of the ATP-dependent chaperone activity of HSP70 provokes reversible cytoplasmic TDP-43 de-mixing and transition from liquid to gel/solid, independently of RNA binding or stress granules. Isotope labelling mass spectrometry was used to identify that phase-separated cytoplasmic TDP-43 is bound by the small heat-shock protein HSPB1. Binding is direct, mediated through TDP-43's RNA binding and low-complexity domains. HSPB1 partitions into TDP-43 droplets, inhibits TDP-43 assembly into fibrils, and is essential for disassembly of stress-induced TDP-43 droplets. A decrease in HSPB1 promotes cytoplasmic TDP-43 de-mixing and mislocalization. HSPB1 depletion was identified in spinal motor neurons of patients with ALS containing aggregated TDP-43. These findings identify HSPB1 to be a regulator of cytoplasmic TDP-43 phase separation and aggregation.


Sujet(s)
Protéines de liaison à l'ADN , Petites protéines du choc thermique , Protéines du choc thermique , Transition de phase , Adénosine triphosphate , Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/métabolisme , Protéines de liaison à l'ADN/composition chimique , Protéines de liaison à l'ADN/métabolisme , Protéines du choc thermique HSP70/métabolisme , Protéines du choc thermique/composition chimique , Protéines du choc thermique/métabolisme , Humains , Chaperons moléculaires/génétique , Proteasome endopeptidase complex , ARN/métabolisme
10.
Cell Rep ; 40(3): 111086, 2022 07 19.
Article de Anglais | MEDLINE | ID: mdl-35858576

RÉSUMÉ

Hyperosmotic stress as physiologic dysfunction can reduce the cell volume and then redistribute both protein concentration and ionic strength, but its effect on liquid-liquid phase separation (LLPS) is not well understood. Here, we map the hyperosmotic-stress-induced nuclear LLPS of amyotrophic lateral sclerosis (ALS)-related proteins (fused in sarcoma [FUS], TAR DNA-binding protein 43 [TDP-43]). The dynamic and reversibility of FUS granules are continuable with the increase of hypertonic stimulation time, but those of TDP-43 granules decrease significantly. Strikingly, FUS granules, but not TDP-43 granules, contain essential chaperone Hsp40, which can protect amyloid protein from solid aggregation. Moreover, FUS nuclear granules can co-localize with paraspeckles, but not promyelocytic leukemia (PML) bodies or nuclear speckles, while TDP-43 nuclear granules cannot co-localize with the above nuclear bodies. Together, these results may broaden our understanding of the LLPS of ALS-related proteins in response to cellular stress.


Sujet(s)
Sclérose latérale amyotrophique , Sclérose latérale amyotrophique/métabolisme , Noyau de la cellule/métabolisme , Humains , Protéine FUS de liaison à l'ARN/métabolisme
11.
iScience ; 25(6): 104356, 2022 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-35620440

RÉSUMÉ

Hsp70 is a key molecular chaperone in the protein quality control system to safeguard protein homeostasis in cells. Previous studies have shown that Hsp70 chaperones TDP-43, a pathogenic protein associated with amyotrophic lateral sclerosis (ALS), in nuclear bodies and prevents it from the pathological aggregation. In this work, we report that Hsp70 undergoes liquid-liquid phase separation, chaperones FUS, another ALS-linked pathogenic protein, in stress granules (SGs), and prevents condensed FUS from amyloid aggregation. Knock-down of Hsp70 does not influence SG assembly but results in the liquid-to-solid transition in SGs. NMR experiments further reveal Hsp70 predominantly uses its C-terminal substrate-binding domain to interact with the low complexity domain of FUS, which represents a mechanism distinct from that interacting with TDP-43. These findings suggest that Hsp70 is widely involved in chaperoning the physiological dynamics of various membrane-less organelles and adopts different mechanisms to prevent the pathological aggregation of different proteins.

12.
Protein Cell ; 13(8): 602-614, 2022 08.
Article de Anglais | MEDLINE | ID: mdl-35384603

RÉSUMÉ

The nucleocapsid (N) protein of SARS-CoV-2 has been reported to have a high ability of liquid-liquid phase separation, which enables its incorporation into stress granules (SGs) of host cells. However, whether SG invasion by N protein occurs in the scenario of SARS-CoV-2 infection is unknow, neither do we know its consequence. Here, we used SARS-CoV-2 to infect mammalian cells and observed the incorporation of N protein into SGs, which resulted in markedly impaired self-disassembly but stimulated cell cellular clearance of SGs. NMR experiments further showed that N protein binds to the SG-related amyloid proteins via non-specific transient interactions, which not only expedites the phase transition of these proteins to aberrant amyloid aggregation in vitro, but also promotes the aggregation of FUS with ALS-associated P525L mutation in cells. In addition, we found that ACE2 is not necessary for the infection of SARS-CoV-2 to mammalian cells. Our work indicates that SARS-CoV-2 infection can impair the disassembly of host SGs and promote the aggregation of SG-related amyloid proteins, which may lead to an increased risk of neurodegeneration.


Sujet(s)
Sclérose latérale amyotrophique , COVID-19 , Protéines amyloïdogènes/métabolisme , Sclérose latérale amyotrophique/génétique , Animaux , Granulations cytoplasmiques/métabolisme , Mammifères , SARS-CoV-2 , Granules de stress
13.
Dev Cell ; 57(5): 583-597.e6, 2022 03 14.
Article de Anglais | MEDLINE | ID: mdl-35231447

RÉSUMÉ

As sessile organisms, plants are highly sensitive to environmental stresses. In response to stresses, globally repressed translation initiation leads to stress granule (SG) formation. Protein liquid-liquid phase separation (LLPS) contributes to SG formation, but a direct link between protein LLPS and stress resistance has not yet been found in plants. Here, we report that two RNA-binding proteins, RBGD2 and RBGD4, function redundantly to improve heat resistance in Arabidopsis. RBGD2 and RBGD4 undergo LLPS in vitro and condense into heat-induced SGs in vivo via tyrosine residue array (TRA). Importantly, disrupting LLPS by mutating TRA abolishes RBGD2/4 condensation in SGs and impairs their protective function against heat stress (HS). Further study found that upon HS, the RBGD2/4 interaction network expands with additional SG proteins and heat-responsive mRNA. Our work shows a mechanistic basis that underlies protein LLPS in HS response in plants and suggests manipulation of protein LLPS as a general strategy to improve plant stress resistance.


Sujet(s)
Arabidopsis , Arabidopsis/génétique , Arabidopsis/métabolisme , Granulations cytoplasmiques/métabolisme , Réaction de choc thermique , ARN messager/génétique , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Stress physiologique
14.
FASEB J ; 36(3): e22084, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35107844

RÉSUMÉ

Chronic kidney disease (CKD), a disease involving damage to the kidney structure and function, is a global public health problem. Tubulointerstitial fibrosis (TIF) is both an inevitable pathological change in individuals with CKD and a driving force in the progression of renal fibrosis. Nicotinamide N-methyltransferase (NNMT) and its metabolite 1-methylnicotinamide (MNAM) have been shown to protect against lipotoxicity-induced kidney tubular injury. However, the biological roles of NNMT and MNAM in regulating TIF remain elusive. This study aimed to investigate the protective effect of NNMT and MNAM on TIF and the mechanisms involved. We explored the functions and mechanisms of NNMT and MNAM in TIF, as well as the interaction between NNMT and MNAM, using unilateral ureteral obstruction (UUO) mice and cultured mouse tubular epithelial cells (mTECs) stimulated with transforming growth factor-ß1 (TGF-ß1). Several important findings were obtained as follows: (1) NNMT expression was upregulated in the kidneys of UUO mice and TGF-ß1-induced mTECs, and this upregulation was proposed to be a protective compensatory response to TIF. (2) MNAM was a potentially effective antifibrotic and anti-inflammatory medication in UUO mice. (3) The antifibrotic effect of NNMT overexpression was exerted by increasing the concentration of MNAM. (4) The renoprotective role of MNAM depended on the selective blockade of the interaction of Smad3 with TGFß receptor I. Overall, our study shows that NNMT is involved in the development and progression of CKD and that its metabolite MNAM may be a novel inhibitor of the TGF-ß1/Smad3 pathway with great therapeutic potential for CKD.


Sujet(s)
Fibrose/métabolisme , Nicotinamide/analogues et dérivés , Nicotinamide N-methyltransferase/métabolisme , Insuffisance rénale chronique/métabolisme , Transduction du signal/physiologie , Protéine Smad-3/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Animaux , Cellules cultivées , Rein/métabolisme , Mâle , Souris , Souris de lignée C57BL , Nicotinamide/métabolisme , Obstruction urétérale/métabolisme
15.
iScience ; 25(1): 103701, 2022 Jan 21.
Article de Anglais | MEDLINE | ID: mdl-35036880

RÉSUMÉ

FUS is a multifunctional nuclear protein which undergoes liquid-liquid phase separation in response to stress and DNA damage. Dysregulation of FUS dynamic phase separation leads to formation of pathological fibril closely associated with neurodegenerative diseases such as amyotrophic lateral sclerosis and frontotemporal dementia. In this study, we determined the cryo-EM structure of a cytotoxic fibril formed by the low-complexity (LC) domain of FUS at 2.9 Å resolution. The fibril structure exhibits a new and extensive serpentine fold consisting of three motifs incorporating together via a Tyr triad. FUS LC employs 91 residues to form an enlarged and stable fibril core via hydrophilic interaction and hydrogen bonds, which is distinct from most of previously determined fibrils commonly stabilized by hydrophobic interaction. Our work reveals the structural basis underlying formation of a cytotoxic and thermostable fibril of FUS LC and sheds light on understanding the liquid-to-solid phase transition of FUS in disease.

17.
Nat Commun ; 11(1): 6349, 2020 12 11.
Article de Anglais | MEDLINE | ID: mdl-33311513

RÉSUMÉ

Human heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) serves as a key regulating protein in RNA metabolism. Malfunction of hnRNPA1 in nucleo-cytoplasmic transport or dynamic phase separation leads to abnormal amyloid aggregation and neurodegeneration. The low complexity (LC) domain of hnRNPA1 drives both dynamic phase separation and amyloid aggregation. Here, we use cryo-electron microscopy to determine the amyloid fibril structure formed by hnRNPA1 LC domain. Remarkably, the structure reveals that the nuclear localization sequence of hnRNPA1 (termed PY-NLS), which is initially known to mediate the nucleo-cytoplamic transport of hnRNPA1 through binding with karyopherin-ß2 (Kapß2), represents the major component of the fibril core. The residues that contribute to the binding of PY-NLS with Kapß2 also exert key molecular interactions to stabilize the fibril structure. Notably, hnRNPA1 mutations found in familial amyotrophic lateral sclerosis (ALS) and multisystem proteinopathoy (MSP) are all involved in the fibril core and contribute to fibril stability. Our work illuminates structural understandings of the pathological amyloid aggregation of hnRNPA1 and the amyloid disaggregase activity of Kapß2, and highlights the multiple roles of PY-NLS in hnRNPA1 homeostasis.


Sujet(s)
Amyloïde/métabolisme , Noyau de la cellule/métabolisme , Ribonucléoprotéine nucléaire hétérogène A1/composition chimique , Ribonucléoprotéine nucléaire hétérogène A1/génétique , Ribonucléoprotéine nucléaire hétérogène A1/métabolisme , Transport nucléaire actif , Sclérose latérale amyotrophique , Cryomicroscopie électronique , Cellules HEK293 , Humains , Caryophérines/métabolisme , Modèles moléculaires , Mutation , Conformation des protéines , Domaines protéiques
18.
Proc Natl Acad Sci U S A ; 117(49): 31123-31133, 2020 12 08.
Article de Anglais | MEDLINE | ID: mdl-33229560

RÉSUMÉ

Membraneless organelles contain a wide spectrum of molecular chaperones, indicating their important roles in modulating the metastable conformation and biological function of membraneless organelles. Here we report that class I and II Hsp40 (DNAJ) proteins possess a high ability of phase separation rendered by the flexible G/F-rich region. Different Hsp40 proteins localize in different membraneless organelles. Specifically, human Hdj1 (DNAJB1), a class II Hsp40 protein, condenses in ubiquitin (Ub)-rich nuclear bodies, while Hdj2 (DNAJA1), a class I Hsp40 protein, condenses in nucleoli. Upon stress, both Hsp40 proteins incorporate into stress granules (SGs). Mutations of the G/F-rich region not only markedly impaired Hdj1 phase separation and SG involvement and disrupted the synergistic phase separation and colocalization of Hdj1 and fused in sarcoma (FUS) in cells. Being cophase separated with FUS, Hdj1 stabilized the liquid phase of FUS against proceeding into amyloid aggregation in vitro and alleviated abnormal FUS aggregation in cells. Moreover, Hdj1 uses different domains to chaperone FUS phase separation and amyloid aggregation. This paper suggests that phase separation is an intrinsic property of Hsp40 proteins, which enables efficient incorporation and function of Hsp40 in membraneless organelles and may further mediate the buildup of chaperone network in membraneless organelles.


Sujet(s)
Protéines du choc thermique HSP40/génétique , Chaperons moléculaires/génétique , Membrane cellulaire/génétique , Humains , Membranes , Organites/génétique , Liaison aux protéines/génétique , Protéine FUS de liaison à l'ARN/génétique , Ubiquitine/génétique
19.
Mol Cell ; 79(3): 443-458.e7, 2020 08 06.
Article de Anglais | MEDLINE | ID: mdl-32649883

RÉSUMÉ

Despite the prominent role of TDP-43 in neurodegeneration, its physiological and pathological functions are not fully understood. Here, we report an unexpected role of TDP-43 in the formation of dynamic, reversible, liquid droplet-like nuclear bodies (NBs) in response to stress. Formation of NBs alleviates TDP-43-mediated cytotoxicity in mammalian cells and fly neurons. Super-resolution microscopy reveals distinct functions of the two RRMs in TDP-43 NB formation. TDP-43 NBs are partially colocalized with nuclear paraspeckles, whose scaffolding lncRNA NEAT1 is dramatically upregulated in stressed neurons. Moreover, increase of NEAT1 promotes TDP-43 liquid-liquid phase separation (LLPS) in vitro. Finally, we discover that the ALS-associated mutation D169G impairs the NEAT1-mediated TDP-43 LLPS and NB assembly, causing excessive cytoplasmic translocation of TDP-43 to form stress granules, which become phosphorylated TDP-43 cytoplasmic foci upon prolonged stress. Together, our findings suggest a stress-mitigating role and mechanism of TDP-43 NBs, whose dysfunction may be involved in ALS pathogenesis.


Sujet(s)
Sclérose latérale amyotrophique/génétique , Protéines de liaison à l'ADN/génétique , Corps d'inclusion intranucléaire/métabolisme , Neurones/métabolisme , ARN long non codant/génétique , Sclérose latérale amyotrophique/métabolisme , Sclérose latérale amyotrophique/anatomopathologie , Animaux , Animal génétiquement modifié , Arsénites/pharmacologie , Cortex cérébral/effets des médicaments et des substances chimiques , Cortex cérébral/métabolisme , Cortex cérébral/ultrastructure , Granulations cytoplasmiques/effets des médicaments et des substances chimiques , Granulations cytoplasmiques/métabolisme , Granulations cytoplasmiques/ultrastructure , Protéines de liaison à l'ADN/métabolisme , Modèles animaux de maladie humaine , Drosophila melanogaster , Régulation de l'expression des gènes , Cellules HEK293 , Cellules HeLa , Humains , Corps d'inclusion intranucléaire/effets des médicaments et des substances chimiques , Corps d'inclusion intranucléaire/ultrastructure , Souris , Mutation , Neurones/effets des médicaments et des substances chimiques , Neurones/ultrastructure , Culture de cellules primaires , Transport des protéines/effets des médicaments et des substances chimiques , ARN long non codant/métabolisme , Transduction du signal , Stress physiologique
20.
Front Chem ; 8: 489, 2020.
Article de Anglais | MEDLINE | ID: mdl-32596208

RÉSUMÉ

The most severe problem in bone regeneration is the defect in the interface. We prepared four types of implantation scaffolds of crosslinked gelatin (GE)/hydroxyapatite (HAp) to study the factors influencing interface interactions, they are film-crosslinked GE scaffold, gel-crosslinked GE scaffold, solid-crosslinked GE/HAp scaffold and gel-crosslinked GE/HAp scaffold. HAp could penetrate the entire GE matrix completely in four successive steps: physical preparation of a gel; chemical crosslinking; incubation in modified simulated body fluid (m-SBF) and freeze-drying. The penetrative nucleation and growth of HAp and the influencing factors in the GE matrix were investigated to ameliorate the interface interactions between organic and inorganic layers. During development of penetrative nucleation and growth, a tight connection was built between organic and inorganic layers, B-type carbonated HAp was formed after incubation with m-BSF, and the apatite content could be controlled. In summary, enhanced interface relies on not only the pre-seeded hydroxyapatite (HAp) as crystal nuclei but also the sufficient space for ions with high concentration to diffuse in.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...