Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.167
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 173(1): 62-73.e9, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29526462

RESUMEN

Aggregates of human islet amyloid polypeptide (IAPP) in the pancreas of patients with type 2 diabetes (T2D) are thought to contribute to ß cell dysfunction and death. To understand how IAPP harms cells and how this might be overcome, we created a yeast model of IAPP toxicity. Ste24, an evolutionarily conserved protease that was recently reported to degrade peptides stuck within the translocon between the cytoplasm and the endoplasmic reticulum, was the strongest suppressor of IAPP toxicity. By testing variants of the human homolog, ZMPSTE24, with varying activity levels, the rescue of IAPP toxicity proved to be directly proportional to the declogging efficiency. Clinically relevant ZMPSTE24 variants identified in the largest database of exomes sequences derived from T2D patients were characterized using the yeast model, revealing 14 partial loss-of-function variants, which were enriched among diabetes patients over 2-fold. Thus, clogging of the translocon by IAPP oligomers may contribute to ß cell failure.


Asunto(s)
Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/química , Polipéptido Amiloide de los Islotes Pancreáticos/toxicidad , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Metaloendopeptidasas/química , Metaloendopeptidasas/genética , Modelos Biológicos , Mutagénesis , Agregado de Proteínas/fisiología , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos
2.
Mol Cell ; 73(5): 1028-1043.e5, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30733118

RESUMEN

Mutations in PTEN-induced kinase 1 (PINK1) can cause recessive early-onset Parkinson's disease (PD). Import arrest results in PINK1 kinase activation specifically on damaged mitochondria, triggering Parkin-mediated mitophagy. Here, we show that PINK1 import is less dependent on Tim23 than on mitochondrial membrane potential (ΔΨm). We identified a negatively charged amino acid cluster motif that is evolutionarily conserved just C-terminal to the PINK1 transmembrane. PINK1 that fails to accumulate at the outer mitochondrial membrane, either by mutagenesis of this negatively charged motif or by deletion of Tom7, is imported into depolarized mitochondria and cleaved by the OMA1 protease. Some PD patient mutations also are defective in import arrest and are rescued by the suppression of OMA1, providing a new potential druggable target for PD. These results suggest that ΔΨm loss-dependent PINK1 import arrest does not result solely from Tim23 inactivation but also through an actively regulated "tug of war" between Tom7 and OMA1.


Asunto(s)
Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Mitocondrias/enzimología , Membranas Mitocondriales/enzimología , Proteínas Mitocondriales/metabolismo , Enfermedad de Parkinson/enzimología , Proteínas Quinasas/metabolismo , Secuencias de Aminoácidos , Antiparkinsonianos/farmacología , Transporte Biológico , Diseño de Fármacos , Activación Enzimática , Células HeLa , Humanos , Potencial de la Membrana Mitocondrial , Proteínas de la Membrana/genética , Metaloendopeptidasas/genética , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Membranas Mitocondriales/efectos de los fármacos , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteínas Mitocondriales/genética , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , Dominios y Motivos de Interacción de Proteínas , Proteínas Quinasas/genética , Proteolisis , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Mol Microbiol ; 121(5): 1063-1078, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38558112

RESUMEN

Metalloprotease-gp63 is a virulence factor secreted by Leishmania. However, secretory pathway in Leishmania is not well defined. Here, we cloned and expressed the GRASP homolog from Leishmania. We found that Leishmania expresses one GRASP homolog of 58 kDa protein (LdGRASP) which localizes in LdRab1- and LPG2-positive Golgi compartment in Leishmania. LdGRASP was found to bind with COPII complex, LdARF1, LdRab1 and LdRab11 indicating its role in ER and Golgi transport in Leishmania. To determine the function of LdGRASP, we generated LdGRASP knockout parasites using CRISPR-Cas9. We found fragmentation of Golgi in Ld:GRASPKO parasites. Our results showed enhanced transport of non-GPI-anchored gp63 to the cell surface leading to higher secretion of this form of gp63 in Ld:GRASPKO parasites in comparison to Ld:WT cells. In contrast, we found that transport of GPI-anchored gp63 to the cell surface is blocked in Ld:GRASPKO parasites and thereby inhibits its secretion. The overexpression of dominant-negative mutant of LdRab1 or LdSar1 in Ld:GRASPKO parasites significantly blocked the secretion of non-GPI-anchored gp63. Interestingly, we found that survival of transgenic parasites overexpressing Ld:GRASP-GFP is significantly compromised in macrophages in comparison to Ld:WT and Ld:GRASPKO parasites. These results demonstrated that LdGRASP differentially regulates Ldgp63 secretory pathway in Leishmania.


Asunto(s)
Metaloendopeptidasas , Proteínas Protozoarias , Factores de Virulencia , Factores de Virulencia/metabolismo , Factores de Virulencia/genética , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/genética , Metaloendopeptidasas/metabolismo , Metaloendopeptidasas/genética , Aparato de Golgi/metabolismo , Retículo Endoplásmico/metabolismo , Macrófagos/parasitología , Macrófagos/metabolismo , Animales , Leishmania/metabolismo , Leishmania/genética , Transporte de Proteínas , Sistemas CRISPR-Cas , Proteínas de la Matriz de Golgi/metabolismo , Proteínas de la Matriz de Golgi/genética
4.
Nature ; 575(7782): 361-365, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31695197

RESUMEN

Reprogramming of mitochondria provides cells with the metabolic flexibility required to adapt to various developmental transitions such as stem cell activation or immune cell reprogramming, and to respond to environmental challenges such as those encountered under hypoxic conditions or during tumorigenesis1-3. Here we show that the i-AAA protease YME1L rewires the proteome of pre-existing mitochondria in response to hypoxia or nutrient starvation. Inhibition of mTORC1 induces a lipid signalling cascade via the phosphatidic acid phosphatase LIPIN1, which decreases phosphatidylethanolamine levels in mitochondrial membranes and promotes proteolysis. YME1L degrades mitochondrial protein translocases, lipid transfer proteins and metabolic enzymes to acutely limit mitochondrial biogenesis and support cell growth. YME1L-mediated mitochondrial reshaping supports the growth of pancreatic ductal adenocarcinoma (PDAC) cells as spheroids or xenografts. Similar changes to the mitochondrial proteome occur in the tumour tissues of patients with PDAC, suggesting that YME1L is relevant to the pathophysiology of these tumours. Our results identify the mTORC1-LIPIN1-YME1L axis as a post-translational regulator of mitochondrial proteostasis at the interface between metabolism and mitochondrial dynamics.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Metabolismo de los Lípidos , Metaloendopeptidasas/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/genética , Hipoxia de la Célula , Línea Celular , Proliferación Celular , Humanos , Lípidos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Metaloendopeptidasas/genética , Proteínas Mitocondriales/genética , Proteolisis
5.
Proc Natl Acad Sci U S A ; 119(40): e2123030119, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36161902

RESUMEN

Lorises are a group of globally threatened strepsirrhine primates that exhibit many unusual physiological and behavioral features, including a low metabolic rate, slow movement, and hibernation. Here, we assembled a chromosome-level genome sequence of the pygmy loris (Xanthonycticebus pygmaeus) and resequenced whole genomes from 50 pygmy lorises and 6 Bengal slow lorises (Nycticebus bengalensis). We found that many gene families involved in detoxification have been specifically expanded in the pygmy loris, including the GSTA gene family, with many newly derived copies functioning specifically in the liver. We detected many genes displaying evolutionary convergence between pygmy loris and koala, including PITRM1. Significant decreases in PITRM1 enzymatic activity in these two species may have contributed to their characteristic low rate of metabolism. We also detected many evolutionarily convergent genes and positively selected genes in the pygmy loris that are involved in muscle development. Functional assays demonstrated the decreased ability of one positively selected gene, MYOF, to up-regulate the fast-type muscle fiber, consistent with the lower proportion of fast-twitch muscle fibers in the pygmy loris. The protein product of another positively selected gene in the pygmy loris, PER2, exhibited weaker binding to the key circadian core protein CRY, a finding that may be related to this species' unusual circadian rhythm. Finally, population genomics analysis revealed that these two extant loris species, which coexist in the same habitat, have exhibited an inverse relationship in terms of their demography over the past 1 million years, implying strong interspecies competition after speciation.


Asunto(s)
Adaptación Biológica , Evolución Biológica , Lorisidae , Adaptación Biológica/genética , Animales , Demografía , Hibernación , Lorisidae/genética , Metagenómica , Metaloendopeptidasas/genética
6.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35197292

RESUMEN

Prelamin A is a farnesylated precursor of lamin A, a nuclear lamina protein. Accumulation of the farnesylated prelamin A variant progerin, with an internal deletion including its processing site, causes Hutchinson-Gilford progeria syndrome. Loss-of-function mutations in ZMPSTE24, which encodes the prelamin A processing enzyme, lead to accumulation of full-length farnesylated prelamin A and cause related progeroid disorders. Some data suggest that prelamin A also accumulates with physiological aging. Zmpste24-/- mice die young, at ∼20 wk. Because ZMPSTE24 has functions in addition to prelamin A processing, we generated a mouse model to examine effects solely due to the presence of permanently farnesylated prelamin A. These mice have an L648R amino acid substitution in prelamin A that blocks ZMPSTE24-catalyzed processing to lamin A. The LmnaL648R/L648R mice express only prelamin and no mature protein. Notably, nearly all survive to 65 to 70 wk, with ∼40% of male and 75% of female LmnaL648R/L648R mice having near-normal lifespans of 90 wk (almost 2 y). Starting at ∼10 wk of age, LmnaL648R/L648R mice of both sexes have lower body masses than controls. By ∼20 to 30 wk of age, they exhibit detectable cranial, mandibular, and dental defects similar to those observed in Zmpste24-/- mice and have decreased vertebral bone density compared to age- and sex-matched controls. Cultured embryonic fibroblasts from LmnaL648R/L648R mice have aberrant nuclear morphology that is reversible by treatment with a protein farnesyltransferase inhibitor. These novel mice provide a model to study the effects of farnesylated prelamin A during physiological aging.


Asunto(s)
Lamina Tipo A/metabolismo , Longevidad , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Progeria/genética , Animales , Sitios de Unión , Proteínas de la Membrana/genética , Metaloendopeptidasas/genética , Ratones , Mutación , Fenotipo , Prenilación
7.
EMBO J ; 39(24): e105074, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33200421

RESUMEN

The connectivity of mitochondria is regulated by a balance between fusion and division. Many human diseases are associated with excessive mitochondrial connectivity due to impaired Drp1, a dynamin-related GTPase that mediates division. Here, we report a mitochondrial stress response, named mitochondrial safeguard, that adjusts the balance of fusion and division in response to increased mitochondrial connectivity. In cells lacking Drp1, mitochondria undergo hyperfusion. However, hyperfusion does not completely connect mitochondria because Opa1 and mitofusin 1, two other dynamin-related GTPases that mediate fusion, become proteolytically inactivated. Pharmacological and genetic experiments show that the activity of Oma1, a metalloprotease that cleaves Opa1, is regulated by short pulses of the membrane depolarization without affecting the overall membrane potential in Drp1-knockout cells. Re-activation of Opa1 and Mitofusin 1 in Drp1-knockout cells further connects mitochondria beyond hyperfusion, termed extreme fusion, leading to bioenergetic deficits. These findings reveal an unforeseen safeguard mechanism that prevents extreme fusion of mitochondria, thereby maintaining mitochondrial function when the balance is shifted to excessive connectivity.


Asunto(s)
Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Mitocondrias/metabolismo , Estrés Fisiológico/fisiología , Animales , Dinaminas/genética , Dinaminas/metabolismo , Metabolismo Energético , GTP Fosfohidrolasas/metabolismo , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Metaloproteasas/metabolismo , Ratones , Mitocondrias/genética , Dinámicas Mitocondriales/fisiología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/metabolismo , Estrés Fisiológico/genética , Transcriptoma
8.
Biochem Biophys Res Commun ; 712-713: 149961, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38648679

RESUMEN

Blood pressure is a crucial physiological parameter and its abnormalities can cause a variety of health problems. We have previously reported that mice with systemic deletion of nardilysin (NRDC), an M16 family metalloprotease, exhibit hypotension. In this study, we aimed to clarify the role of NRDC in vascular smooth muscle cell (VSMC) by generating VSMC-specific Nrdc knockout (VSMC-KO) mice. Our findings reveal that VSMC-KO mice also exhibit hypotension. Aortas isolated from VSMC-KO mice exhibited a weakened contractile response to phenylephrine, accompanied by reduced phosphorylation of myosin light chain 2 and decreased rhoA expression. VSMC isolated from VSMC-KO aortas showed a reduced increase in intracellular Ca2+ concentration induced by α-stimulants. These findings suggest that NRDC in VSMC regulates vascular contraction and blood pressure by modulating Ca2+ dynamics.


Asunto(s)
Presión Sanguínea , Calcio , Metaloendopeptidasas , Ratones Noqueados , Músculo Liso Vascular , Miocitos del Músculo Liso , Animales , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Calcio/metabolismo , Ratones , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Metaloendopeptidasas/metabolismo , Metaloendopeptidasas/genética , Masculino , Ratones Endogámicos C57BL , Hipotensión/metabolismo , Células Cultivadas , Aorta/metabolismo , Aorta/citología , Vasoconstricción/efectos de los fármacos , Señalización del Calcio
9.
BMC Plant Biol ; 24(1): 265, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38600480

RESUMEN

BACKGROUND: Leaf variegation is an intriguing phenomenon observed in many plant species. However, questions remain on its mechanisms causing patterns of different colours. In this study, we describe a tomato plant detected in an M2 population of EMS mutagenised seeds, showing variegated leaves with sectors of dark green (DG), medium green (MG), light green (LG) hues, and white (WH). Cells and tissues of these classes, along with wild-type tomato plants, were studied by light, fluorescence, and transmission electron microscopy. We also measured chlorophyll a/b and carotene and quantified the variegation patterns with a machine-learning image analysis tool. We compared the genomes of pooled plants with wild-type-like and mutant phenotypes in a segregating F2 population to reveal candidate genes responsible for the variegation. RESULTS: A genetic test demonstrated a recessive nuclear mutation caused the variegated phenotype. Cross-sections displayed distinct anatomy of four-leaf phenotypes, suggesting a stepwise mesophyll degradation. DG sectors showed large spongy layers, MG presented intercellular spaces in palisade layers, and LG displayed deformed palisade cells. Electron photomicrographs of those mesophyll cells demonstrated a gradual breakdown of the chloroplasts. Chlorophyll a/b and carotene were proportionally reduced in the sectors with reduced green pigments, whereas white sectors have hardly any of these pigments. The colour segmentation system based on machine-learning image analysis was able to convert leaf variegation patterns into binary images for quantitative measurements. The bulk segregant analysis of pooled wild-type-like and variegated progeny enabled the identification of SNP and InDels via bioinformatic analysis. The mutation mapping bioinformatic pipeline revealed a region with three candidate genes in chromosome 4, of which the FtsH-like protein precursor (LOC100037730) carries an SNP that we consider the causal variegated phenotype mutation. Phylogenetic analysis shows the candidate is evolutionary closest to the Arabidopsis VAR1. The synonymous mutation created by the SNP generated a miRNA binding site, potentially disrupting the photoprotection mechanism and thylakoid development, resulting in leaf variegation. CONCLUSION: We described the histology, anatomy, physiology, and image analysis of four classes of cell layers and chloroplast degradation in a tomato plant with a variegated phenotype. The genomics and bioinformatics pipeline revealed a VAR1-related FtsH mutant, the first of its kind in tomato variegation phenotypes. The miRNA binding site of the mutated SNP opens the way to future studies on its epigenetic mechanism underlying the variegation.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , MicroARNs , Solanum lycopersicum , Solanum lycopersicum/genética , Clorofila A/metabolismo , Filogenia , Cloroplastos/genética , Arabidopsis/genética , Mutación , Fenotipo , Hojas de la Planta/metabolismo , Carotenoides/metabolismo , MicroARNs/metabolismo , Precursores de Proteínas/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Proteínas de Arabidopsis/genética
10.
Pediatr Res ; 95(5): 1356-1362, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38191824

RESUMEN

BACKGROUND: Hutchinson-Gilford progeria syndrome (HGPS) and progeroid laminopathies (PL) are extremely rare genetic diseases with extremely poor prognoses. This study aims to investigate the epidemiological and genotypic characteristics of patients with HGPS/PL in China. METHODS: Using a cross-sectional study design, general characteristics and genotypic data of 46 patients with HGPS/PL from 17 provinces in China were analyzed. RESULTS: Among the 46 patients with HGPS/PL, 20 patients are HGPS, and the rest are PL; the identified total prevalence of HGPS/PL is 1/23 million. Among 42 patients with gene reports, 3 carried compound heterozygous mutations in the ZMPSTE24 while the other 39 carried LMNA mutations. Among PL, LMNA c.1579 C > T homozygous mutation was the most common. The onset of classic genotype HGPS is skin sclerosis in the first month after birth. The primary clinical manifestations of PL patients include skin abnormalities, growth retardation, and joint stiffness. The median age of onset for PL was 12 (6,12) months. CONCLUSIONS: In China, the identified total prevalence of HGPS/PL is 1/23 million. 92.8% of the genetic mutations of HGPS/PL were located in LMNA, and the rest in ZMPSTE24. Most patients of HGPS/PL have skin abnormalities as the earliest manifestation. Compared to PL, the classic genotype HGPS starts earlier. IMPACT STATEMENT: Hutchinson-Gilford progeria syndrome (HGPS) and progeroid laminopathies (PL) are extremely rare genetic diseases with extremely poor prognoses. To date, there is a paucity of epidemiological data related to HGPS/PL in China. This study first examined the genotypic, phenotypic, and prevalence characteristics of 40-50% of the cases of HGPS/PL in mainland China through a collaborative international registry effort. In China, the identified total prevalence of HGPS/PL is 1/23 million. 92.8% of the genetic mutations of HGPS/PL are located in LMNA. LMNA c.1579 C > T homozygous mutations are the most common form of gene mutations among the Chinese PL population.


Asunto(s)
Lamina Tipo A , Proteínas de la Membrana , Mutación , Progeria , Humanos , Progeria/genética , Progeria/epidemiología , China/epidemiología , Masculino , Femenino , Lamina Tipo A/genética , Estudios Transversales , Preescolar , Lactante , Prevalencia , Niño , Proteínas de la Membrana/genética , Metaloendopeptidasas/genética , Genotipo , Adolescente , Laminopatías/genética , Laminopatías/epidemiología , Fenotipo
11.
J Peripher Nerv Syst ; 29(2): 262-274, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38860315

RESUMEN

BACKGROUND: Loss-of-function variants in MME (membrane metalloendopeptidase) are a known cause of recessive Charcot-Marie-Tooth Neuropathy (CMT). A deep intronic variant, MME c.1188+428A>G (NM_000902.5), was identified through whole genome sequencing (WGS) of two Australian families with recessive inheritance of axonal CMT using the seqr platform. MME c.1188+428A>G was detected in a homozygous state in Family 1, and in a compound heterozygous state with a known pathogenic MME variant (c.467del; p.Pro156Leufs*14) in Family 2. AIMS: We aimed to determine the pathogenicity of the MME c.1188+428A>G variant through segregation and splicing analysis. METHODS: The splicing impact of the deep intronic MME variant c.1188+428A>G was assessed using an in vitro exon-trapping assay. RESULTS: The exon-trapping assay demonstrated that the MME c.1188+428A>G variant created a novel splice donor site resulting in the inclusion of an 83 bp pseudoexon between MME exons 12 and 13. The incorporation of the pseudoexon into MME transcript is predicted to lead to a coding frameshift and premature termination codon (PTC) in MME exon 14 (p.Ala397ProfsTer47). This PTC is likely to result in nonsense mediated decay (NMD) of MME transcript leading to a pathogenic loss-of-function. INTERPRETATION: To our knowledge, this is the first report of a pathogenic deep intronic MME variant causing CMT. This is of significance as deep intronic variants are missed using whole exome sequencing screening methods. Individuals with CMT should be reassessed for deep intronic variants, with splicing impacts being considered in relation to the potential pathogenicity of variants.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth , Intrones , Linaje , Empalme del ARN , Humanos , Enfermedad de Charcot-Marie-Tooth/genética , Masculino , Femenino , Empalme del ARN/genética , Intrones/genética , Metaloendopeptidasas/genética , Adulto , Mutación
12.
Brain ; 146(10): 4117-4131, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37086482

RESUMEN

Hereditary spastic paraplegia is a neurological condition characterized by predominant axonal degeneration in long spinal tracts, leading to weakness and spasticity in the lower limbs. The nicotinamide adenine dinucleotide (NAD+)-consuming enzyme SARM1 has emerged as a key executioner of axonal degeneration upon nerve transection and in some neuropathies. An increase in the nicotinamide mononucleotide/NAD+ ratio activates SARM1, causing catastrophic NAD+ depletion and axonal degeneration. However, the role of SARM1 in the pathogenesis of hereditary spastic paraplegia has not been investigated. Here, we report an enhanced mouse model for hereditary spastic paraplegia caused by mutations in SPG7. The eSpg7 knockout mouse carries a deletion in both Spg7 and Afg3l1, a redundant homologue expressed in mice but not in humans. The eSpg7 knockout mice recapitulate the phenotypic features of human patients, showing progressive symptoms of spastic-ataxia and degeneration of axons in the spinal cord as well as the cerebellum. We show that the lack of SPG7 rewires the mitochondrial proteome in both tissues, leading to an early onset decrease in mito-ribosomal subunits and a remodelling of mitochondrial solute carriers and transporters. To interrogate mechanisms leading to axonal degeneration in this mouse model, we explored the involvement of SARM1. Deletion of SARM1 delays the appearance of ataxic signs, rescues mitochondrial swelling and axonal degeneration of cerebellar granule cells and dampens neuroinflammation in the cerebellum. The loss of SARM1 also prevents endoplasmic reticulum abnormalities in long spinal cord axons, but does not halt the degeneration of these axons. Our data thus reveal a neuron-specific interplay between SARM1 and mitochondrial dysfunction caused by lack of SPG7 in hereditary spastic paraplegia.


Asunto(s)
Paraplejía Espástica Hereditaria , Animales , Humanos , Ratones , Proteínas del Dominio Armadillo/genética , ATPasas Asociadas con Actividades Celulares Diversas , Axones/patología , Cerebelo , Proteínas del Citoesqueleto/genética , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , NAD , Paraplejía Espástica Hereditaria/genética
13.
Mol Cell ; 64(1): 148-162, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27642048

RESUMEN

Mutations in subunits of mitochondrial m-AAA proteases in the inner membrane cause neurodegeneration in spinocerebellar ataxia (SCA28) and hereditary spastic paraplegia (HSP7). m-AAA proteases preserve mitochondrial proteostasis, mitochondrial morphology, and efficient OXPHOS activity, but the cause for neuronal loss in disease is unknown. We have determined the neuronal interactome of m-AAA proteases in mice and identified a complex with C2ORF47 (termed MAIP1), which counteracts cell death by regulating the assembly of the mitochondrial Ca2+ uniporter MCU. While MAIP1 assists biogenesis of the MCU subunit EMRE, the m-AAA protease degrades non-assembled EMRE and ensures efficient assembly of gatekeeper subunits with MCU. Loss of the m-AAA protease results in accumulation of constitutively active MCU-EMRE channels lacking gatekeeper subunits in neuronal mitochondria and facilitates mitochondrial Ca2+ overload, mitochondrial permeability transition pore opening, and neuronal death. Together, our results explain neuronal loss in m-AAA protease deficiency by deregulated mitochondrial Ca2+ homeostasis.


Asunto(s)
Canales de Calcio/metabolismo , Cerebelo/metabolismo , Cuerpo Estriado/metabolismo , Hipocampo/metabolismo , Metaloendopeptidasas/genética , Mitocondrias/metabolismo , Neuronas/metabolismo , Proteasas ATP-Dependientes/genética , Proteasas ATP-Dependientes/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Animales , Calcio/metabolismo , Canales de Calcio/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Muerte Celular , Cerebelo/patología , Cuerpo Estriado/patología , Regulación de la Expresión Génica , Células HEK293 , Hipocampo/patología , Homeostasis/genética , Humanos , Transporte Iónico , Metaloendopeptidasas/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/patología , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Neuronas/patología , Mapeo de Interacción de Proteínas , Transducción de Señal
14.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33658366

RESUMEN

A challenge faced by peptidases is the recognition of highly diverse substrates. A feature of some peptidase families is the capacity to specifically use post-translationally added glycans present on their protein substrates as a recognition determinant. This is ultimately critical to enabling peptide bond hydrolysis. This class of enzyme is also frequently large and architecturally sophisticated. However, the molecular details underpinning glycan recognition by these O-glycopeptidases, the importance of these interactions, and the functional roles of their ancillary domains remain unclear. Here, using the Clostridium perfringens ZmpA, ZmpB, and ZmpC M60 peptidases as model proteins, we provide structural and functional insight into how these intricate proteins recognize glycans as part of catalytic and noncatalytic substrate recognition. Structural, kinetic, and mutagenic analyses support the key role of glycan recognition within the M60 domain catalytic site, though they point to ZmpA as an apparently inactive enzyme. Wider examination of the Zmp domain content reveals noncatalytic carbohydrate binding as a feature of these proteins. The complete three-dimensional structure of ZmpB provides rare insight into the overall molecular organization of a highly multimodular enzyme and reveals how the interplay of individual domain function may influence biological activity. O-glycopeptidases frequently occur in host-adapted microbes that inhabit or attack mucus layers. Therefore, we anticipate that these results will be fundamental to informing more detailed models of how the glycoproteins that are abundant in mucus are destroyed as part of pathogenic processes or liberated as energy sources during normal commensal lifestyles.


Asunto(s)
Proteínas Bacterianas/química , Clostridium perfringens/enzimología , Metaloendopeptidasas/química , Mucinas/química , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/química , Proteínas Bacterianas/genética , Dominio Catalítico , Clostridium perfringens/genética , Hidrólisis , Metaloendopeptidasas/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética
15.
Br J Cancer ; 128(11): 1991-1999, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36991255

RESUMEN

BACKGROUND: Prostate cancer is the most common cancer in men in the developed world, with most deaths caused by advanced and metastatic disease which has no curative options. Here, we identified Mbtps2 alteration to be associated with metastatic disease in an unbiased in vivo screen and demonstrated its regulation of fatty acid and cholesterol metabolism. METHODS: The Sleeping Beauty transposon system was used to randomly alter gene expression in the PtenNull murine prostate. MBTPS2 was knocked down by siRNA in LNCaP, DU145 and PC3 cell lines, which were then phenotypically investigated. RNA-Seq was performed on LNCaP cells lacking MBTPS2, and pathways validated by qPCR. Cholesterol metabolism was investigated by Filipin III staining. RESULTS: Mbtps2 was identified in our transposon-mediated in vivo screen to be associated with metastatic prostate cancer. Silencing of MBTPS2 expression in LNCaP, DU145 and PC3 human prostate cancer cells reduced proliferation and colony forming growth in vitro. Knockdown of MBTPS2 expression in LNCaP cells impaired cholesterol synthesis and uptake along with reduced expression of key regulators of fatty acid synthesis, namely FASN and ACACA. CONCLUSION: MBTPS2 is implicated in progressive prostate cancer and may mechanistically involve its effects on fatty acid and cholesterol metabolism.


Asunto(s)
Lipogénesis , Neoplasias de la Próstata , Masculino , Humanos , Animales , Ratones , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Línea Celular Tumoral , Neoplasias de la Próstata/patología , Colesterol , Ácidos Grasos , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo
16.
Nat Immunol ; 12(1): 45-53, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21151101

RESUMEN

Cytotoxic T lymphocytes (CTLs) recognize peptides presented by HLA class I molecules on the cell surface. The C terminus of these CTL epitopes is considered to be produced by the proteasome. Here we demonstrate that the cytosolic endopeptidases nardilysin and thimet oligopeptidase (TOP) complemented proteasome activity. Nardilysin and TOP were required, either together or alone, for the generation of a tumor-specific CTL epitope from PRAME, an immunodominant CTL epitope from Epstein-Barr virus protein EBNA3C, and a clinically important epitope from the melanoma protein MART-1. TOP functioned as C-terminal trimming peptidase in antigen processing, and nardilysin contributed to both the C-terminal and N-terminal generation of CTL epitopes. By broadening the antigenic peptide repertoire, nardilysin and TOP strengthen the immune defense against intracellular pathogens and cancer.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Epítopos de Linfocito T/metabolismo , Metaloendopeptidasas/metabolismo , Linfocitos T Citotóxicos/metabolismo , Presentación de Antígeno/genética , Antígenos de Neoplasias/química , Antígenos de Neoplasias/inmunología , Citotoxicidad Inmunológica/genética , Epítopos de Linfocito T/química , Epítopos de Linfocito T/inmunología , Antígeno HLA-A3/metabolismo , Humanos , Células K562 , Metaloendopeptidasas/genética , Metaloendopeptidasas/inmunología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , ARN Interferente Pequeño/genética , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Transgenes/genética
17.
Clin Genet ; 104(4): 491-496, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37270786

RESUMEN

Restrictive dermopathy (RD) is a lethal condition caused by biallelic loss-of-function mutations in ZMPSTE24, whereas mutations preserving residual enzymatic activity of the ZMPSTE24 protein lead to the milder mandibuloacral dysplasia with type B lipodystrophy (MADB) phenotype. Remarkably, we identified a homozygous, presumably loss-of-function mutation in ZMPSTE24 [c.28_29insA, p.(Leu10Tyrfs*37)] in two consanguineous Pakistani families segregating MADB. To clarify how lethal consequences are prevented in affected individuals, functional analysis was performed. Expression experiments supported utilization of two alternative translation initiation sites, preventing complete loss of protein function consistent with the relatively mild phenotypic outcome in affected patients. One of these alternative start codons is newly formed at the insertion site. Our findings indicate that the creation of new potential start codons through N-terminal mutations in other disease-associated genes should generally be taken into consideration in the variant interpretation process.


Asunto(s)
Mutación del Sistema de Lectura , Metaloendopeptidasas , Humanos , Mutación del Sistema de Lectura/genética , Codón Iniciador/genética , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Mutación , Codón , Proteínas de la Membrana/genética
18.
Am J Med Genet A ; 191(2): 582-585, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36367250

RESUMEN

Spastic paraplegia is a neurodegenerative disorder characterized by progressive leg weakness and spasticity due to degeneration of corticospinal axons. SPG7 encodes paraplegin, and pathogenic variants in the gene cause hereditary spastic paraplegia as an autosomal recessive trait. Various ophthalmological findings including optic atrophy, ophthalmoplegia, or nystagmus have been reported in patients with spastic paraplegia type 7. We report a 15-year-old male patient with a novel heterozygous variant, c.1224T>G:p.(Asp408Glu) in SPG7 (NM_003119.3) causing early onset isolated optic atrophy and infantile nystagmus prior to the onset of neurological symptoms. Therefore, SPG7 should be considered a cause of infantile nystagmus with optic atrophy.


Asunto(s)
Atrofia Óptica Autosómica Dominante , Atrofia Óptica , Paraplejía Espástica Hereditaria , Humanos , Masculino , ATPasas Asociadas con Actividades Celulares Diversas/genética , Metaloendopeptidasas/genética , Mutación , Atrofia Óptica/diagnóstico , Atrofia Óptica/genética , Atrofia Óptica/patología , Paraplejía/genética , Fenotipo , Paraplejía Espástica Hereditaria/complicaciones , Paraplejía Espástica Hereditaria/diagnóstico , Paraplejía Espástica Hereditaria/genética , Adolescente
19.
Mol Cell ; 60(1): 47-62, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26387735

RESUMEN

Mitochondrial permeability transition is a phenomenon in which the mitochondrial permeability transition pore (PTP) abruptly opens, resulting in mitochondrial membrane potential (ΔΨm) dissipation, loss of ATP production, and cell death. Several genetic candidates have been proposed to form the PTP complex, however, the core component is unknown. We identified a necessary and conserved role for spastic paraplegia 7 (SPG7) in Ca(2+)- and ROS-induced PTP opening using RNAi-based screening. Loss of SPG7 resulted in higher mitochondrial Ca(2+) retention, similar to cyclophilin D (CypD, PPIF) knockdown with sustained ΔΨm during both Ca(2+) and ROS stress. Biochemical analyses revealed that the PTP is a heterooligomeric complex composed of VDAC, SPG7, and CypD. Silencing or disruption of SPG7-CypD binding prevented Ca(2+)- and ROS-induced ΔΨm depolarization and cell death. This study identifies an ubiquitously expressed IMM integral protein, SPG7, as a core component of the PTP at the OMM and IMM contact site.


Asunto(s)
Ciclofilinas/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Mitocondrias/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Sitios de Unión , Calcio/metabolismo , Muerte Celular , Ciclofilinas/química , Células HEK293 , Células HeLa , Humanos , Potencial de la Membrana Mitocondrial , Metaloendopeptidasas/química , Membranas Mitocondriales/metabolismo , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo
20.
Neurol Sci ; 44(9): 3303-3305, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37213040

RESUMEN

BACKGROUND: Progressive muscular atrophy (PMA) is a rare adult-onset neurological disease that is characterized by isolated lower motor neuron degeneration. While it is still disputable whether PMA is a subtype of amyotrophic lateral sclerosis (ALS) or an isolated disorder, it is well-established as a clinically defined entity. About 5% of PMA cases are monogenic, and the implicated genes largely overlap with those causing monogenic ALS. CASE DESCRIPTION: Here we describe a 68-year-old female patient with progressive and asymmetric upper-limb weakness throughout an 18-month period, with muscle atrophy, dysphagia and slurring of speech. The lower limbs were unaffected, and there was no sign of upper motor neuron dysfunction. Comprehensive genetic testing for single nucleotide and copy-number variants revealed a pathogenic monoallelic variant c.1529C>T, p.(Ala510Val) in the SPG7 gene. DISCUSSION: Pathogenic biallelic SPG7 variants have been originally associated with hereditary spastic paraplegia, but other phenotypes are nowadays known to be linked to these variants, such as ALS. However, there is no report of this (or any) other SPG7 variant in association with PMA, whether it progressed to ALS or not. In conclusion, we present the first known case of PMA associated with a monoallelic SPG7 mutation.


Asunto(s)
Esclerosis Amiotrófica Lateral , Atrofia Muscular Espinal , Paraplejía Espástica Hereditaria , Femenino , Humanos , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Atrofia Muscular Espinal/genética , Mutación/genética , Pruebas Genéticas , Paraplejía Espástica Hereditaria/genética , ATPasas Asociadas con Actividades Celulares Diversas/genética , Metaloendopeptidasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA