Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 335
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(40): e2318098121, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39331414

RESUMEN

Huntington disease (HD) is a genetic neurodegenerative disease caused by cytosine, adenine, guanine (CAG) expansion in the Huntingtin (HTT) gene, translating to an expanded polyglutamine tract in the HTT protein. Age at disease onset correlates to CAG repeat length but varies by decades between individuals with identical repeat lengths. Genome-wide association studies link HD modification to DNA repair and mitochondrial health pathways. Clinical studies show elevated DNA damage in HD, even at the premanifest stage. A major DNA repair node influencing neurodegenerative disease is the PARP pathway. Accumulation of poly adenosine diphosphate (ADP)-ribose (PAR) has been implicated in Alzheimer and Parkinson diseases, as well as cerebellar ataxia. We report that HD mutation carriers have lower cerebrospinal fluid PAR levels than healthy controls, starting at the premanifest stage. Human HD induced pluripotent stem cell-derived neurons and patient-derived fibroblasts have diminished PAR response in the context of elevated DNA damage. We have defined a PAR-binding motif in HTT, detected HTT complexed with PARylated proteins in human cells during stress, and localized HTT to mitotic chromosomes upon inhibition of PAR degradation. Direct HTT PAR binding was measured by fluorescence polarization and visualized by atomic force microscopy at the single molecule level. While wild-type and mutant HTT did not differ in their PAR binding ability, purified wild-type HTT protein increased in vitro PARP1 activity while mutant HTT did not. These results provide insight into an early molecular mechanism of HD, suggesting possible targets for the design of early preventive therapies.


Asunto(s)
Proteína Huntingtina , Enfermedad de Huntington , Poli Adenosina Difosfato Ribosa , Transducción de Señal , Humanos , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Proteína Huntingtina/metabolismo , Proteína Huntingtina/genética , Poli Adenosina Difosfato Ribosa/metabolismo , Daño del ADN , Neuronas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Fibroblastos/metabolismo , Reparación del ADN
2.
AAPS J ; 26(6): 106, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39289253

RESUMEN

To evaluate the artificial intelligence (AI)-guided AlphaFold algorithm for studying the binding interactions of human huntingtin and the aggregation of huntingtin peptides. Variants of huntingtin protein implicated in Huntington's disease were used as a model system to evaluate AlphaFold. Variants of huntingtin and huntingtin peptides with polyglutamine tracts (PQT) containing 21, 31, 51, or 78 glutamines were studied. The 3-dimensional structures of huntingtin variants and their interactions with huntingtin-associated protein-40 (HAP40) were obtained. Aggregation experiments were conducted with peptide sequences corresponding to variants of PQT, amino terminal sequence (NTS) plus PQT, NTS plus PQT plus proline rich region (PRR), and the 300 amino acid sequence from the NTS through HEAT3 of huntingtin. Oligomerization experiments with 1, 3, 6, or 12 peptide sequences were used to assess the quaternary structures of aggregates. The PQT and PQT plus NTS peptides formed a helical secondary structure that formed a central core in the quaternary structure of the aggregates The PRR formed an extended type II polyproline helix that did not participate in central core the aggregates. The distance between the amino and carboxyl termini of disease-linked 31Q, 51Q, and 78Q variants of full-length huntingtin was prominently decreased compared to the 21Q huntingtin. The interaction of HAP40 with the 78Q variant increased the distance between the amino and carboxyl termini. AlphaFold identified key tertiary structure changes in human huntingtin that have been independently corroborated in experimental models. The results highlight the utility of AlphaFold for hypothesis generation in pharmaceutical research.


Asunto(s)
Proteína Huntingtina , Enfermedad de Huntington , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/metabolismo , Humanos , Farmacogenética/métodos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Inteligencia Artificial , Algoritmos , Péptidos/genética , Péptidos/química , Modelos Moleculares , Secuencia de Aminoácidos
3.
J Cell Biol ; 223(11)2024 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-39150509

RESUMEN

Huntington's disease (HD) is caused by a polyglutamine expansion of the huntingtin protein, resulting in the formation of polyglutamine aggregates. The mechanisms of toxicity that result in the complex HD pathology remain only partially understood. Here, we show that nuclear polyglutamine aggregates induce nuclear envelope (NE) blebbing and ruptures that are often repaired incompletely. These ruptures coincide with disruptions of the nuclear lamina and lead to lamina scar formation. Expansion microscopy enabled resolving the ultrastructure of nuclear aggregates and revealed polyglutamine fibrils sticking into the cytosol at rupture sites, suggesting a mechanism for incomplete repair. Furthermore, we found that NE repair factors often accumulated near nuclear aggregates, consistent with stalled repair. These findings implicate nuclear polyQ aggregate-induced loss of NE integrity as a potential contributing factor to Huntington's disease and other polyglutamine diseases.


Asunto(s)
Enfermedad de Huntington , Membrana Nuclear , Péptidos , Membrana Nuclear/metabolismo , Membrana Nuclear/ultraestructura , Humanos , Péptidos/metabolismo , Péptidos/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Enfermedad de Huntington/genética , Animales , Proteína Huntingtina/metabolismo , Proteína Huntingtina/genética , Agregado de Proteínas , Lámina Nuclear/metabolismo , Lámina Nuclear/ultraestructura , Núcleo Celular/metabolismo
4.
IUCrJ ; 11(Pt 5): 849-858, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39120045

RESUMEN

The aberrant fibrillization of huntingtin exon 1 (Httex1) characterized by an expanded polyglutamine (polyQ) tract is a defining feature of Huntington's disease, a neurodegenerative disorder. Recent investigations underscore the involvement of a small EDRK-rich factor 1a (SERF1a) in promoting Httex1 fibrillization through interactions with its N terminus. By establishing an integrated approach with size-exclusion-column-based small- and wide-angle X-ray scattering (SEC-SWAXS), NMR, and molecular simulations using Rosetta, the analysis here reveals a tight binding of two NT17 fragments of Httex1 (comprising the initial 17 amino acids at the N terminus) to the N-terminal region of SERF1a. In contrast, examination of the complex structure of SERF1a with a coiled NT17-polyQ peptide (33 amino acids in total) indicates sparse contacts of the NT17 and polyQ segments with the N-terminal side of SERF1a. Furthermore, the integrated SEC-SWAXS and molecular-simulation analysis suggests that the coiled NT17 segment can transform into a helical conformation when associated with a polyQ segment exhibiting high helical content. Intriguingly, NT17-polyQ peptides with enhanced secondary structures display diminished interactions with SERF1a. This insight into the conformation-dependent binding of NT17 provides clues to a catalytic association mechanism underlying SERF1a's facilitation of Httext1 fibrillization.


Asunto(s)
Proteína Huntingtina , Péptidos , Proteína Huntingtina/genética , Proteína Huntingtina/química , Proteína Huntingtina/metabolismo , Péptidos/química , Péptidos/metabolismo , Péptidos/genética , Humanos , Exones/genética , Unión Proteica , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Simulación de Dinámica Molecular , Espectroscopía de Resonancia Magnética , Difracción de Rayos X
5.
J Phys Chem B ; 128(32): 7761-7769, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39092631

RESUMEN

Huntington's disease (HD) is a fatal neurodegenerative disorder resulting from an abnormal expansion of polyglutamine (polyQ) repeats in the N-terminus of the huntingtin protein. When the polyQ tract surpasses 35 repeats, the mutated protein undergoes misfolding, culminating in the formation of intracellular aggregates. Research in mouse models suggests that HD pathogenesis involves the aggregation of N-terminal fragments of the huntingtin protein (htt). These early oligomeric assemblies of htt, exhibiting diverse characteristics during aggregation, are implicated as potential toxic entities in HD. However, a consensus on their specific structures remains elusive. Understanding the heterogeneous nature of htt oligomers provides crucial insights into disease mechanisms, emphasizing the need to identify various oligomeric conformations as potential therapeutic targets. Employing coarse-grained molecular dynamics, our study aims to elucidate the mechanisms governing the aggregation process and resultant aggregate architectures of htt. The polyQ tract within htt is flanked by two regions: an N-terminal domain (N17) and a short C-terminal proline-rich segment. We conducted self-assembly simulations involving five distinct N17 + polyQ systems with polyQ lengths ranging from 7 to 45, utilizing the ProMPT force field. Prolongation of the polyQ domain correlates with an increase in ß-sheet-rich structures. Longer polyQ lengths favor intramolecular ß-sheets over intermolecular interactions due to the folding of the elongated polyQ domain into hairpin-rich conformations. Importantly, variations in polyQ length significantly influence resulting oligomeric structures. Shorter polyQ domains lead to N17 domain aggregation, forming a hydrophobic core, while longer polyQ lengths introduce a competition between N17 hydrophobic interactions and polyQ polar interactions, resulting in densely packed polyQ cores with outwardly distributed N17 domains. Additionally, at extended polyQ lengths, we observe distinct oligomeric conformations with varying degrees of N17 bundling. These findings can help explain the toxic gain-of-function that htt with expanded polyQ acquires.


Asunto(s)
Proteína Huntingtina , Simulación de Dinámica Molecular , Péptidos , Proteína Huntingtina/química , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Péptidos/química , Péptidos/metabolismo , Humanos , Multimerización de Proteína , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/genética
6.
Int J Mol Sci ; 25(14)2024 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-39062952

RESUMEN

Gastrodin (GAS) is the main chemical component of the traditional Chinese herb Gastrodia elata (called "Tianma" in Chinese), which has been used to treat neurological conditions, including headaches, epilepsy, stroke, and memory loss. To our knowledge, it is unclear whether GAS has a therapeutic effect on Huntington's disease (HD). In the present study, we evaluated the effect of GAS on the degradation of mutant huntingtin protein (mHtt) by using PC12 cells transfected with N-terminal mHtt Q74. We found that 0.1-100 µM GAS had no effect on the survival rate of Q23 and Q74 PC12 cells after 24-48 h of incubation. The ubiquitin-proteasome system (UPS) is the main system that clears misfolded proteins in eukaryotic cells. Mutated Htt significantly upregulated total ubiquitinated protein (Ub) expression, decreased chymotrypsin-like, trypsin-like and caspase-like peptidase activity, and reduced the colocalization of the 20S proteasome with mHtt. GAS (25 µM) attenuated all of the abovementioned pathological changes, and the regulatory effect of GAS on mHtt was found to be abolished by MG132, a proteasome inhibitor. The autophagy-lysosome pathway (ALP) is another system for misfolded protein degradation. Although GAS downregulated the expression of autophagy markers (LC3II and P62), it increased the colocalization of LC3II with lysosomal associated membrane protein 1 (LAMP1), which indicates that ALP was activated. Moreover, GAS prevented mHtt-induced neuronal damage in PC12 cells. GAS has a selective effect on mHtt in Q74 PC12 cells and has no effect on Q23 and proteins encoded by other genes containing long CAGs, such as Rbm33 (10 CAG repeats) and Hcn1 (>30 CAG repeats). Furthermore, oral administration of 100 mg/kg GAS increased grip strength and attenuated mHtt aggregates in B6-hHTT130-N transgenic mice. This is a high dose (100 mg/kg GAS) when compared with experiments on HD mice with other small molecules. We will design more doses to evaluate the dose-response relationship of the inhibition effect of GAS on mHtt in our next study. In summary, GAS can promote the degradation of mHtt by activating the UPS and ALP, making it a potential therapeutic agent for HD.


Asunto(s)
Autofagia , Alcoholes Bencílicos , Glucósidos , Proteína Huntingtina , Lisosomas , Complejo de la Endopetidasa Proteasomal , Ubiquitina , Animales , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Ratas , Complejo de la Endopetidasa Proteasomal/metabolismo , Células PC12 , Autofagia/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/efectos de los fármacos , Ubiquitina/metabolismo , Alcoholes Bencílicos/farmacología , Glucósidos/farmacología , Ratones , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/genética , Proteolisis/efectos de los fármacos , Mutación
7.
Proc Natl Acad Sci U S A ; 121(32): e2319091121, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39074279

RESUMEN

Understanding the normal function of the Huntingtin (HTT) protein is of significance in the design and implementation of therapeutic strategies for Huntington's disease (HD). Expansion of the CAG repeat in the HTT gene, encoding an expanded polyglutamine (polyQ) repeat within the HTT protein, causes HD and may compromise HTT's normal activity contributing to HD pathology. Here, we investigated the previously defined role of HTT in autophagy specifically through studying HTT's association with ubiquitin. We find that HTT interacts directly with ubiquitin in vitro. Tandem affinity purification was used to identify ubiquitinated and ubiquitin-associated proteins that copurify with a HTT N-terminal fragment under basal conditions. Copurification is enhanced by HTT polyQ expansion and reduced by mimicking HTT serine 421 phosphorylation. The identified HTT-interacting proteins include RNA-binding proteins (RBPs) involved in mRNA translation, proteins enriched in stress granules, the nuclear proteome, the defective ribosomal products (DRiPs) proteome and the brain-derived autophagosomal proteome. To determine whether the proteins interacting with HTT are autophagic targets, HTT knockout (KO) cells and immunoprecipitation of lysosomes were used to investigate autophagy in the absence of HTT. HTT KO was associated with reduced abundance of mitochondrial proteins in the lysosome, indicating a potential compromise in basal mitophagy, and increased lysosomal abundance of RBPs which may result from compensatory up-regulation of starvation-induced macroautophagy. We suggest HTT is critical for appropriate basal clearance of mitochondrial proteins and RBPs, hence reduced HTT proteostatic function with mutation may contribute to the neuropathology of HD.


Asunto(s)
Proteína Huntingtina , Lisosomas , Mitocondrias , Proteínas de Unión al ARN , Ubiquitina , Proteína Huntingtina/metabolismo , Proteína Huntingtina/genética , Lisosomas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Humanos , Ubiquitina/metabolismo , Mitocondrias/metabolismo , Autofagia , Animales , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Ratones , Unión Proteica , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Péptidos/metabolismo
8.
Life Sci Alliance ; 7(9)2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39054288

RESUMEN

Huntington's disease (HD) is a fatal neurodegenerative disorder caused by an expanded CAG tract in the huntingtin (HTT) gene, leading to toxic gains of function. HTT-lowering treatments are in clinical trials, but the risks imposed are unclear. Recent studies have reported on the consequences of widespread HTT loss in mice, where one group described early HTT loss leading to fatal pancreatitis, but later loss as benign. Another group reported no pancreatitis but found widespread neurological phenotypes including subcortical calcification. To better understand the liabilities of widespread HTT loss, we knocked out Htt with two separate tamoxifen-inducible Cre lines. We find that loss of HTT at 2 mo of age leads to progressive tremors and severe subcortical calcification at examination at 14 mo of age but does not result in acute pancreatitis or histological changes in the pancreas. We, in addition, report that HTT loss is followed by sustained induction of circulating neurofilament light chain. These results confirm that global loss of HTT in mice is associated with pronounced risks, including progressive subcortical calcification and neurodegeneration.


Asunto(s)
Modelos Animales de Enfermedad , Proteína Huntingtina , Enfermedad de Huntington , Ratones Noqueados , Páncreas , Animales , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Ratones , Páncreas/patología , Páncreas/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Enfermedad de Huntington/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/patología , Masculino , Calcinosis/genética , Calcinosis/patología , Fenotipo , Femenino
9.
J Phys Chem Lett ; 15(24): 6375-6382, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38857530

RESUMEN

The effects of two macromolecular cosolutes, specifically the polysaccharide dextran-20 and the protein lysozyme, on the aggregation kinetics of a pathogenic huntingtin exon-1 protein (hhtex1) with a 35 polyglutamine repeat, httex1Q35, are described. A unified kinetic model that establishes a direct connection between reversible tetramerization occurring on the microsecond time scale and irreversible fibril formation on a time scale of hours/days forms the basis for quantitative analysis of httex1Q35 aggregation, monitored by measuring cross-peak intensities in a series of 2D 1H-15N NMR correlation spectra acquired during the course of aggregation. The primary effects of the two cosolutes are associated with shifts in the prenucleation tetramerization equilibrium resulting in substantial changes in concentration of "preformed" httex1Q35 tetramers. Similar effects of the two cosolutes on the tetramerization equilibrium observed for a shorter, nonaggregating huntingtin variant with a 7-glutamine repeat, httex1Q7, lend confidence to the conclusions drawn from the fits to the httex1Q35 aggregation kinetics.


Asunto(s)
Proteína Huntingtina , Muramidasa , Proteína Huntingtina/química , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Cinética , Muramidasa/química , Muramidasa/metabolismo , Humanos , Dextranos/química , Péptidos/química , Resonancia Magnética Nuclear Biomolecular , Agregado de Proteínas/efectos de los fármacos , Sustancias Macromoleculares/química , Multimerización de Proteína/efectos de los fármacos , Espectroscopía de Resonancia Magnética
10.
Cells ; 13(12)2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38920658

RESUMEN

The development of cell-type-specific dendritic arbors is integral to the proper functioning of neurons within their circuit networks. In this study, we examine the regulatory relationship between the cytosolic chaperonin CCT, key insulin pathway genes, and an E3 ubiquitin ligase (Cullin1) in dendritic development. CCT loss of function (LOF) results in dendritic hypotrophy in Drosophila Class IV (CIV) multi-dendritic larval sensory neurons, and CCT has recently been shown to fold components of the TOR (Target of Rapamycin) complex 1 (TORC1) in vitro. Through targeted genetic manipulations, we confirm that an LOF of CCT and the TORC1 pathway reduces dendritic complexity, while overexpression of key TORC1 pathway genes increases the dendritic complexity in CIV neurons. Furthermore, both CCT and TORC1 LOF significantly reduce microtubule (MT) stability. CCT has been previously implicated in regulating proteinopathic aggregation, thus, we examine CIV dendritic development in disease conditions as well. The expression of mutant Huntingtin leads to dendritic hypotrophy in a repeat-length-dependent manner, which can be rescued by Cullin1 LOF. Together, our data suggest that Cullin1 and CCT influence dendritic arborization through the regulation of TORC1 in both health and disease.


Asunto(s)
Proteínas Cullin , Dendritas , Proteínas de Drosophila , Drosophila melanogaster , Animales , Proteínas Cullin/metabolismo , Proteínas Cullin/genética , Dendritas/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteína Huntingtina/metabolismo , Proteína Huntingtina/genética , Larva/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Microtúbulos/metabolismo , Células Receptoras Sensoriales/metabolismo , Transducción de Señal , Factores de Transcripción , Chaperonina con TCP-1
11.
Am J Hum Genet ; 111(6): 1165-1183, 2024 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-38749429

RESUMEN

The pathological huntingtin (HTT) trinucleotide repeat underlying Huntington disease (HD) continues to expand throughout life. Repeat length correlates both with earlier age at onset (AaO) and faster progression, making slowing its expansion an attractive therapeutic approach. Genome-wide association studies have identified candidate variants associated with altered AaO and progression, with many found in DNA mismatch repair (MMR)-associated genes. We examine whether lowering expression of these genes affects the rate of repeat expansion in human ex vivo models using HD iPSCs and HD iPSC-derived striatal medium spiny neuron-enriched cultures. We have generated a stable CRISPR interference HD iPSC line in which we can specifically and efficiently lower gene expression from a donor carrying over 125 CAG repeats. Lowering expression of each member of the MMR complexes MutS (MSH2, MSH3, and MSH6), MutL (MLH1, PMS1, PMS2, and MLH3), and LIG1 resulted in characteristic MMR deficiencies. Reduced MSH2, MSH3, and MLH1 slowed repeat expansion to the largest degree, while lowering either PMS1, PMS2, or MLH3 slowed it to a lesser degree. These effects were recapitulated in iPSC-derived striatal cultures where MutL factor expression was lowered. CRISPRi-mediated lowering of key MMR factor expression to levels feasibly achievable by current therapeutic approaches was able to effectively slow the expansion of the HTT CAG tract. We highlight members of the MutL family as potential targets to slow pathogenic repeat expansion with the aim to delay onset and progression of HD and potentially other repeat expansion disorders exhibiting somatic instability.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Proteína Huntingtina , Enfermedad de Huntington , Células Madre Pluripotentes Inducidas , Expansión de Repetición de Trinucleótido , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Reparación de la Incompatibilidad de ADN/genética , Células Madre Pluripotentes Inducidas/metabolismo , Expansión de Repetición de Trinucleótido/genética , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Homólogo 1 de la Proteína MutL/genética , Homólogo 1 de la Proteína MutL/metabolismo , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Genes Modificadores , Proteína 3 Homóloga de MutS/genética , Proteína 3 Homóloga de MutS/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas MutL/genética , Proteínas MutL/metabolismo , Sistemas CRISPR-Cas , Estudio de Asociación del Genoma Completo
12.
Nat Cell Biol ; 26(6): 892-902, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38741019

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder caused by expansion of a CAG trinucleotide repeat in the Huntingtin (HTT) gene, encoding a homopolymeric polyglutamine (polyQ) tract. Although mutant HTT (mHTT) protein is known to aggregate, the links between aggregation and neurotoxicity remain unclear. Here we show that both translation and aggregation of wild-type HTT and mHTT are regulated by a stress-responsive upstream open reading frame and that polyQ expansions cause abortive translation termination and release of truncated, aggregation-prone mHTT fragments. Notably, we find that mHTT depletes translation elongation factor eIF5A in brains of symptomatic HD mice and cultured HD cells, leading to pervasive ribosome pausing and collisions. Loss of eIF5A disrupts homeostatic controls and impairs recovery from acute stress. Importantly, drugs that inhibit translation initiation reduce premature termination and mitigate this escalating cascade of ribotoxic stress and dysfunction in HD.


Asunto(s)
Factor 5A Eucariótico de Iniciación de Traducción , Proteína Huntingtina , Enfermedad de Huntington , Factores de Iniciación de Péptidos , Péptidos , Proteostasis , Proteínas de Unión al ARN , Ribosomas , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Animales , Péptidos/metabolismo , Péptidos/genética , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Humanos , Ribosomas/metabolismo , Ribosomas/genética , Factores de Iniciación de Péptidos/metabolismo , Factores de Iniciación de Péptidos/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Ratones , Ratones Transgénicos , Modelos Animales de Enfermedad , Estrés Fisiológico , Encéfalo/metabolismo , Encéfalo/patología , Expansión de Repetición de Trinucleótido/genética
13.
Biochim Biophys Acta Biomembr ; 1866(6): 184339, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38763270

RESUMEN

Huntington's Disease (HD) is caused by an abnormal expansion of the polyglutamine (polyQ) domain within the first exon of the huntingtin protein (htt). This expansion promotes disease-related htt aggregation into amyloid fibrils and the formation of proteinaceous inclusion bodies within neurons. Fibril formation is a complex heterogenous process involving an array of aggregate species such as oligomers, protofibrils, and fibrils. In HD, structural abnormalities of membranes of several organelles develop. In particular, the accumulation of htt fibrils near the endoplasmic reticulum (ER) impinges upon the membrane, resulting in ER damage, altered dynamics, and leakage of Ca2+. Here, the aggregation of htt at a bilayer interface assembled from ER-derived liposomes was investigated, and fibril formation directly on these membranes was enhanced. Based on these observations, simplified model systems were used to investigate mechanisms associated with htt aggregation on ER membranes. As the ER-derived liposome fractions contained residual Ca2+, the role of divalent cations was also investigated. In the absence of lipids, divalent cations had minimal impact on htt structure and aggregation. However, the presence of Ca2+ or Mg2+ played a key role in promoting fibril formation on lipid membranes despite reduced htt insertion into and association with lipid interfaces, suggesting that the ability of divalent cations to promote fibril formation on membranes is mediated by induced changes to the lipid membrane physicochemical properties. With enhanced concentrations of intracellular calcium being a hallmark of HD, the ability of divalent cations to influence htt aggregation at lipid membranes may play a role in aggregation events that lead to organelle abnormalities associated with disease.


Asunto(s)
Amiloide , Calcio , Cationes Bivalentes , Retículo Endoplásmico , Proteína Huntingtina , Enfermedad de Huntington , Liposomas , Retículo Endoplásmico/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Proteína Huntingtina/química , Humanos , Cationes Bivalentes/metabolismo , Calcio/metabolismo , Amiloide/metabolismo , Amiloide/química , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Enfermedad de Huntington/genética , Liposomas/química , Liposomas/metabolismo , Magnesio/metabolismo , Magnesio/química , Péptidos
14.
J Mol Biol ; 436(12): 168607, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38734203

RESUMEN

Polyglutamine (polyQ) sequences undergo repeat-length dependent formation of disease-associated, amyloid-like cross-ß core structures with kinetics and aggregate morphologies often influenced by the flanking sequences. In Huntington's disease (HD), the httNT segment on the polyQ's N-terminal flank enhances aggregation rates by changing amyloid nucleation from a classical homogeneous mechanism to a two-step process requiring an ɑ-helix-rich oligomeric intermediate. A folded, helix-rich httNT tetrameric structure suggested to be this critical intermediate was recently reported. Here we employ single alanine replacements along the httNT sequence to assess this proposed structure and refine the mechanistic model. We find that Ala replacement of hydrophobic residues within simple httNT peptides greatly suppresses helicity, supporting the tetramer model. These same helix-disruptive replacements in the httNT segment of an exon-1 analog greatly reduce aggregation kinetics, suggesting that an ɑ-helix rich multimer - either the tetramer or a larger multimer - plays an on-pathway role in nucleation. Surprisingly, several other Ala replacements actually enhance helicity and/or amyloid aggregation. The spatial localization of these residues on the tetramer surface suggests a self-association interface responsible for formation of the octomers and higher-order multimers most likely required for polyQ amyloid nucleation. Multimer docking of the tetramer, using the protein-protein docking algorithm ClusPro, predicts this symmetric surface to be a viable tetramer dimerization interface. Intriguingly, octomer formation brings the emerging polyQ chains into closer proximity at this tetramer-tetramer interface. Further supporting the potential importance of tetramer super-assembly, computational docking with a known exon-1 aggregation inhibitor predicts ligand contacts with residues at this interface.


Asunto(s)
Amiloide , Exones , Proteína Huntingtina , Multimerización de Proteína , Humanos , Amiloide/química , Amiloide/metabolismo , Proteína Huntingtina/química , Proteína Huntingtina/metabolismo , Proteína Huntingtina/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/genética , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Modelos Moleculares , Péptidos/química , Péptidos/metabolismo , Agregado de Proteínas
15.
EBioMedicine ; 103: 105124, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38701619

RESUMEN

BACKGROUND: PolyQ diseases are autosomal dominant neurodegenerative disorders caused by the expansion of CAG repeats. While of slow progression, these diseases are ultimately fatal and lack effective therapies. METHODS: A high-throughput chemical screen was conducted to identify drugs that lower the toxicity of a protein containing the first exon of Huntington's disease (HD) protein huntingtin (HTT) harbouring 94 glutamines (Htt-Q94). Candidate drugs were tested in a wide range of in vitro and in vivo models of polyQ toxicity. FINDINGS: The chemical screen identified the anti-leprosy drug clofazimine as a hit, which was subsequently validated in several in vitro models. Computational analyses of transcriptional signatures revealed that the effect of clofazimine was due to the stimulation of mitochondrial biogenesis by peroxisome proliferator-activated receptor gamma (PPARγ). In agreement with this, clofazimine rescued mitochondrial dysfunction triggered by Htt-Q94 expression. Importantly, clofazimine also limited polyQ toxicity in developing zebrafish and neuron-specific worm models of polyQ disease. INTERPRETATION: Our results support the potential of repurposing the antimicrobial drug clofazimine for the treatment of polyQ diseases. FUNDING: A full list of funding sources can be found in the acknowledgments section.


Asunto(s)
Clofazimina , Glutamina , Proteína Huntingtina , Enfermedad de Huntington , PPAR gamma , Péptidos , Pez Cebra , Animales , Humanos , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/metabolismo , Clofazimina/farmacología , Modelos Animales de Enfermedad , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/metabolismo , Leprostáticos/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Péptidos/metabolismo , Péptidos/toxicidad , PPAR gamma/metabolismo , PPAR gamma/genética , Glutamina/metabolismo , Glutamina/toxicidad
16.
Mol Cell ; 84(10): 1980-1994.e8, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38759629

RESUMEN

Aggregation of proteins containing expanded polyglutamine (polyQ) repeats is the cytopathologic hallmark of a group of dominantly inherited neurodegenerative diseases, including Huntington's disease (HD). Huntingtin (Htt), the disease protein of HD, forms amyloid-like fibrils by liquid-to-solid phase transition. Macroautophagy has been proposed to clear polyQ aggregates, but the efficiency of aggrephagy is limited. Here, we used cryo-electron tomography to visualize the interactions of autophagosomes with polyQ aggregates in cultured cells in situ. We found that an amorphous aggregate phase exists next to the radially organized polyQ fibrils. Autophagosomes preferentially engulfed this amorphous material, mediated by interactions between the autophagy receptor p62/SQSTM1 and the non-fibrillar aggregate surface. In contrast, amyloid fibrils excluded p62 and evaded clearance, resulting in trapping of autophagic structures. These results suggest that the limited efficiency of autophagy in clearing polyQ aggregates is due to the inability of autophagosomes to interact productively with the non-deformable, fibrillar disease aggregates.


Asunto(s)
Amiloide , Autofagosomas , Autofagia , Proteína Huntingtina , Enfermedad de Huntington , Péptidos , Agregado de Proteínas , Proteína Sequestosoma-1 , Péptidos/metabolismo , Péptidos/química , Péptidos/genética , Humanos , Proteína Huntingtina/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/química , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Proteína Sequestosoma-1/metabolismo , Proteína Sequestosoma-1/genética , Amiloide/metabolismo , Amiloide/química , Amiloide/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Microscopía por Crioelectrón , Animales , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/genética
17.
Biochem Soc Trans ; 52(2): 719-731, 2024 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-38563485

RESUMEN

The aggregation of proteins into amyloid-like fibrils is seen in many neurodegenerative diseases. Recent years have seen much progress in our understanding of these misfolded protein inclusions, thanks to advances in techniques such as solid-state nuclear magnetic resonance (ssNMR) spectroscopy and cryogenic electron microscopy (cryo-EM). However, multiple repeat-expansion-related disorders have presented special challenges to structural elucidation. This review discusses the special role of ssNMR analysis in the study of protein aggregates associated with CAG repeat expansion disorders. In these diseases, the misfolding and aggregation affect mutant proteins with expanded polyglutamine segments. The most common disorder, Huntington's disease (HD), is connected to the mutation of the huntingtin protein. Since the discovery of the genetic causes for HD in the 1990s, steady progress in our understanding of the role of protein aggregation has depended on the integrative and interdisciplinary use of multiple types of structural techniques. The heterogeneous and dynamic features of polyQ protein fibrils, and in particular those formed by huntingtin N-terminal fragments, have made these aggregates into challenging targets for structural analysis. ssNMR has offered unique insights into many aspects of these amyloid-like aggregates. These include the atomic-level structure of the polyglutamine core, but also measurements of dynamics and solvent accessibility of the non-core flanking domains of these fibrils' fuzzy coats. The obtained structural insights shed new light on pathogenic mechanisms behind this and other protein misfolding diseases.


Asunto(s)
Resonancia Magnética Nuclear Biomolecular , Péptidos , Humanos , Amiloide/química , Amiloide/metabolismo , Proteína Huntingtina/química , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/genética , Péptidos/química , Péptidos/metabolismo , Agregado de Proteínas , Pliegue de Proteína
18.
J Huntingtons Dis ; 13(2): 201-214, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38640164

RESUMEN

Background: Huntington's disease is an inheritable autosomal dominant disorder caused by an expanded CAG trinucleotide repeat within the Huntingtin gene, leading to a polyglutamine (polyQ) expansion in the mutant protein. Objective: A potential therapeutic approach for delaying or preventing the onset of the disease involves enhancing the degradation of the aggregation-prone polyQ-expanded N-terminal mutant huntingtin (mHTT) exon1 fragment. A few proteases and peptidases have been identified that are able to cleave polyQ fragments with low efficiency. This study aims to identify a potent polyQ-degrading endopeptidase. Methods: Here we used quenched polyQ peptides to identify a polyQ-degrading endopeptidase. Next we investigated its role on HTT turnover, using purified polyQ-expanded HTT fragments and striatal cells expressing mHTT exon1 peptides. Results: We identified insulin-degrading enzyme (IDE) as a novel endopeptidase for degrading polyQ peptides. IDE was, however, ineffective in reducing purified polyQ-expanded HTT fragments. Similarly, in striatal cells expressing mHTT exon1 peptides, IDE did not enhance mHTT turnover. Conclusions: This study shows that despite IDE's efficiency in degrading polyQ peptides, it does not contribute to the direct degradation of polyQ-expanded mHTT fragments.


Asunto(s)
Proteína Huntingtina , Insulisina , Péptidos , Insulisina/metabolismo , Insulisina/genética , Proteína Huntingtina/metabolismo , Proteína Huntingtina/genética , Péptidos/metabolismo , Humanos , Animales , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/genética , Ratones , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Cuerpo Estriado/metabolismo
19.
Proc Natl Acad Sci U S A ; 121(13): e2313652121, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38498709

RESUMEN

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by an expanded CAG repeat in the huntingtin (HTT) gene. The repeat-expanded HTT encodes a mutated HTT (mHTT), which is known to induce DNA double-strand breaks (DSBs), activation of the cGAS-STING pathway, and apoptosis in HD. However, the mechanism by which mHTT triggers these events is unknown. Here, we show that HTT interacts with both exonuclease 1 (Exo1) and MutLα (MLH1-PMS2), a negative regulator of Exo1. While the HTT-Exo1 interaction suppresses the Exo1-catalyzed DNA end resection during DSB repair, the HTT-MutLα interaction functions to stabilize MLH1. However, mHTT displays a significantly reduced interaction with Exo1 or MutLα, thereby losing the ability to regulate Exo1. Thus, cells expressing mHTT exhibit rapid MLH1 degradation and hyperactive DNA excision, which causes severe DNA damage and cytosolic DNA accumulation. This activates the cGAS-STING pathway to mediate apoptosis. Therefore, we have identified unique functions for both HTT and mHTT in modulating DNA repair and the cGAS-STING pathway-mediated apoptosis by interacting with MLH1. Our work elucidates the mechanism by which mHTT causes HD.


Asunto(s)
Enfermedad de Huntington , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Proteínas Mutantes/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Nucleotidiltransferasas/genética , ADN , Apoptosis/genética , Homólogo 1 de la Proteína MutL/genética
20.
Adv Sci (Weinh) ; 11(24): e2309217, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38476051

RESUMEN

Pathogenic huntingtin exon-1 protein (httex1), characterized by an expanded polyglutamine tract located between the N-terminal amphiphilic region and a C-terminal polyproline-rich domain, forms fibrils that accumulate in neuronal inclusion bodies, and is associated with a fatal, autosomal dominant neurodegenerative condition known as Huntington's disease. Here a complete kinetic model is described for aggregation/fibril formation of a httex1 construct with a 35-residue polyglutamine repeat, httex1Q35. Using exchange NMR spectroscopy, it is previously shown that the reversible formation of a sparsely-populated tetramer of the N-terminal amphiphilic domain of httex1Q35, comprising a D2 symmetric four-helix bundle, occurs on the microsecond time-scale and is a prerequisite for subsequent nucleation and fibril formation on a time scale that is many orders of magnitude slower (hours). Here a unified kinetic model of httex1Q35 aggregation is developed in which fast, reversible tetramerization is directly linked to slow irreversible fibril formation via conversion of pre-equilibrated tetrameric species to "active", chain elongation-capable nuclei by conformational re-arrangement with a finite, monomer-independent rate. The unified model permits global quantitative analysis of reversible tetramerization and irreversible fibril formation from a time series of 1H-15N correlation spectra recorded during the course of httex1Q35 aggregation.


Asunto(s)
Proteína Huntingtina , Enfermedad de Huntington , Proteína Huntingtina/genética , Proteína Huntingtina/química , Proteína Huntingtina/metabolismo , Humanos , Cinética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/genética , Conformación Proteica , Agregado de Proteínas , Espectroscopía de Resonancia Magnética/métodos , Péptidos/química , Péptidos/metabolismo , Péptidos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA